1
|
Zhong YL, Xu CQ, Li J, Liang ZQ, Wang MM, Ma C, Jia CL, Cao YB, Chen J. Mitochondrial dynamics and metabolism in macrophages for cardiovascular disease: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156620. [PMID: 40068296 DOI: 10.1016/j.phymed.2025.156620] [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: 12/16/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Mitochondria regulate macrophage function, affecting cardiovascular diseases like atherosclerosis and heart failure. Their dynamics interact with macrophage cell death mechanisms, including apoptosis and necroptosis. PURPOSE This review explores how mitochondrial dynamics and metabolism influence macrophage inflammation and cell death in CVDs, highlighting therapeutic targets for enhancing macrophage resilience and reducing CVD pathology, while examining molecular pathways and pharmacological agents involved. STUDY DESIGN This is a narrative review that integrates findings from various studies on mitochondrial dynamics and metabolism in macrophages, their interactions with the endoplasmic reticulum (ER) and Golgi apparatus, and their implications for CVDs. The review also considers the potential therapeutic effects of pharmacological agents on these pathways. METHODS The review utilizes a comprehensive literature search to identify relevant studies on mitochondrial dynamics and metabolism in macrophages, their role in CVDs, and the effects of pharmacological agents on these pathways. The selected studies are analyzed and synthesized to provide insights into the complex relationships between mitochondria, the ER, and Golgi apparatus, and their implications for macrophage function and fate. RESULTS The review reveals that mitochondrial metabolism intertwines with cellular architecture and function, particularly through its intricate interactions with the ER and Golgi apparatus. Mitochondrial-associated membranes (MAMs) facilitate Ca2+ transfer from the ER to mitochondria, maintaining mitochondrial homeostasis during ER stress. The Golgi apparatus transports proteins crucial for inflammatory signaling, contributing to immune responses. Inflammation-induced metabolic reprogramming in macrophages, characterized by a shift from oxidative phosphorylation to glycolysis, underscores the multifaceted role of mitochondrial metabolism in regulating immune cell polarization and inflammatory outcomes. Notably, mitochondrial dysfunction, marked by heightened reactive oxygen species generation, fuels inflammatory cascades and promotes cell death, exacerbating CVD pathology. However, pharmacological agents such as Metformin, Nitazoxanide, and Galanin emerge as potential therapeutic modulators of these pathways, offering avenues for mitigating CVD progression. CONCLUSION This review highlights mitochondrial dynamics and metabolism in macrophage inflammation and cell death in CVDs, suggesting therapeutic targets to improve macrophage resilience and reduce pathology, with new pharmacological agents offering treatment opportunities.
Collapse
Affiliation(s)
- Yi-Lang Zhong
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Chen-Qin Xu
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ji Li
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Zhi-Qiang Liang
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Miao-Miao Wang
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Chao Ma
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Cheng-Lin Jia
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yong-Bing Cao
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Jian Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Anhui Province Rural Revitalization Collaborative Technical Service Center, Huangshan University, Huangshan 245041, China; Department of Public Health, International College, Krirk University, Bangkok, Thailand.
| |
Collapse
|
2
|
Fischer FA, Demarco B, Min FCH, Yeap HW, De Nardo D, Chen KW, Bezbradica JS. TBK1 and IKKε prevent premature cell death by limiting the activity of both RIPK1 and NLRP3 death pathways. SCIENCE ADVANCES 2025; 11:eadq1047. [PMID: 40053580 PMCID: PMC11887814 DOI: 10.1126/sciadv.adq1047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
The loss of TBK1, or both TBK1 and the related kinase IKKε, results in uncontrolled cell death-driven inflammation. Here, we show that the pathway leading to cell death depends on the nature of the activating signal. Previous models suggest that in steady state, TBK1/IKKε-deficient cells die slowly and spontaneously predominantly by uncontrolled tumor necrosis factor-RIPK1-driven death. However, upon infection of cells that express the NLRP3 inflammasome, (e.g., macrophages), with pathogens that activate this pathway (e.g., Listeria monocytogenes), TBK1/IKKε-deficient cells die rapidly, prematurely, and exclusively by enhanced NLRP3-driven pyroptosis. Even infection with the RIPK1-activating pathogen, Yersinia pseudotuberculosis, results in enhanced RIPK1-caspase-8 activation and enhanced secondary NLRP3 activation. Mechanistically, TBK1/IKKε control endosomal traffic, and their loss disrupts endosomal homeostasis, thereby signaling cell stress. This results in premature NLRP3 activation even upon sensing "signal 2" alone, without the obligatory "signal 1." Collectively, TBK1/IKKε emerge as a central brake in limiting death-induced inflammation by both RIPK1 and NLRP3 death-inducing pathways.
Collapse
Affiliation(s)
- Fabian A. Fischer
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Benjamin Demarco
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Felicia Chan Hui Min
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hui Wen Yeap
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dominic De Nardo
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kaiwen W. Chen
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | |
Collapse
|
3
|
Pu Z, Li L, Zhang Y, Shui Y, Liu J, Wang X, Jiang X, Zhang L, Yang H. Exploring the therapeutic potential of HAPC in COVID-19-induced acute lung injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156563. [PMID: 40023068 DOI: 10.1016/j.phymed.2025.156563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Acute lung injury (ALI) is one of the critical complications of coronavirus disease 2019 (COVID-19), which significantly impacts the survival of patients. PURPOSE In this study, we screened COVID-19-related target genes and identified and optimized potential drugs targeting these genes for the treatment of COVID-19. STUDY DESIGN In this study, bioinformatic analyses were conducted and subsequently identified and optimized potential drugs targeting these genes for the treatment of COVID-19 were carried out. METHODS Firstly, we analyzed the targets gene in patients with COVID-19 using single-cell data analysis. We performed structural modifications on Chicoric acid (CA) and combined it with hyaluronic acid to enhance the targeted activity towards Cluster of differentiation 44 (CD44). Poly (sodium-p styrenesulfonate) (PSS) was used to form a PSS-coated CA+hyaluronic acid nanocomplex (HA-P). Subsequently, Lactobacillus murinus conidia cell wall (CW) was encapsulated to prepare PSS-coated CA + hyaluronic acid + Lactobacillus murinus conidia cell wall (HAPC) nanocomplexes. RESULTS The expression of APPL1 expression in macrophage of COVID-19 patients was up-regulation. CA was found to bind to the APPL1 protein and inhibit its ubiquitination. HAPC effectively targeted ALI through the highly efficient interaction between CD44 and Hyaluronic acid (HA). HAPC alleviated the symptoms of ALI and restored epithelial function in mice with ALI. HAPC induced the Adaptor protein containing a pH domain, PTB domain and leucine zipper motif 1 (APPL1)/ liver kinase B1 (LKB1)/ AMP-activated protein kinase (AMPK) pathway by inactivating the NOD - like receptor protein 3 (NLRP3) pathway in ALI. CA interacted with the APPL1 protein and prevented its ubiquitination. HAPC facilitated the interaction between APPL1 and LKB1 to induce the AMPK/NLRP3 pathway. It promoted the formation of LKB1 at GLU-67, ARG-72, ARG-314, ASP-316, and GLN-312 and APPL1 at ARG-106, ASP-115, LYS-124, ASN-119, and GLU-120. CONCLUSION Altogether, HAPC nanocomplexes exerted anti-inflammatory effects on ALI by promoting the interaction between APPL1 and LKB1 to induce the AMPK/NLRP3 pathway, and may be one new therapeutic strategie for ALI.
Collapse
Affiliation(s)
- Zhichen Pu
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui 241001, China,; Drug Clinical Evaluation, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, China
| | - Lingling Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, China
| | - Yan Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, China
| | - Yinping Shui
- Wannan Medical College Wuhu 241001, Anhui, PR China
| | - Jun Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, China
| | - Xiaohu Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiaogan Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, China,.
| | - Liqin Zhang
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui 241001, China,; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, China,.
| | - Hui Yang
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wuhu, Anhui 241001, China,; Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, China,; Tissue bank of the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| |
Collapse
|
4
|
Bu WJ, Li SS, Liu C, Wang YH, Lu JR, Dong CR, Zheng DJ, Fan ZY, Yu Y, Zhang W, Bai YL. Nepetin limits NLRP3 inflammasome activation and alleviates NLRP3-driven inflammatory diseases via PINK1-dependent mitophagy. Free Radic Biol Med 2025; 227:420-433. [PMID: 39653129 DOI: 10.1016/j.freeradbiomed.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
The NLRP3 inflammasome plays a pivotal role in the progression of inflammatory diseases. Mitochondrial damage, oxidative stress and mitochondrial DNA (mtDNA) leak are the key upstream factors for NLRP3 inflammasome activation. Nepetin (Nep), a naturally occurring flavonoid found with anti-inflammatory properties; however, whether it can affect the NLRP3 inflammasome activation and its precise anti-inflammatory mechanism remains unclear. In this study, we demonstrated that Nep enhances PINK1-mediated ubiquitin phosphorylation, which promotes mitophagy and subsequently inhibits NLRP3 inflammasome activation and pyroptosis in macrophages. The administration of Nep to macrophages alleviated of mitochondrial damage, reduced mitochondrial superoxide production, restored mitochondrial membrane potential and prevented the mtDNA leakage. These findings provide compelling evidence for the antioxidant effect of Nep. Furthermore, the pivotal function of mitophagy in the NLRP3 inflammasome inhibitory impact of Nep was substantiated through the utilisation of mitophagy inhibitors and siRNA techniques. Notably, Nep increased survival and reduced organ damage in mice with systemic inflammation by inhibiting NLRP3 inflammasome activation. In addition, Nep suppressed NLRP3 inflammasome activation in obese mice, which led to reduced white adipose and liver inflammation, thereby ameliorating insulin resistance. In conclusion, our findings suggest that Nep is a potent NLRP3 inflammasome inhibitor and a promising candidate for the development of anti-inflammatory therapies.
Collapse
Affiliation(s)
- Wen-Jie Bu
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Si-Si Li
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Chang Liu
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Yue-Hua Wang
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Jian-Rong Lu
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Chao-Run Dong
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Dong-Jie Zheng
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Zhe-Yu Fan
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Yi Yu
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China
| | - Wei Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yun-Long Bai
- Department of Pharmacology, SKLFZCD, (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China.
| |
Collapse
|
5
|
Hong C, Wang L, Zhou X, Zou L, Xiang X, Deng H, Li Q, Wu Y, Liu L, Li T. Protective Effects of Mdivi-1 on Cognition Disturbance Following Sepsis in Mice via Alleviating Microglia Activation and Polarization. CNS Neurosci Ther 2025; 31:e70149. [PMID: 39791542 PMCID: PMC11719124 DOI: 10.1111/cns.70149] [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: 05/20/2024] [Revised: 10/09/2024] [Accepted: 10/20/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Neuroinflammation is one of the essential pathogeneses of cognitive damage suffering from sepsis-associated encephalopathy (SAE). Lots of evidences showed the microglia presented mitochondrial fragmentation during SAE. This study investigated the protective effects and novel mechanisms of inhibiting microglia mitochondrial fragmentation via mitochondrial division inhibitor 1 (Mdivi-1) on cognitive damage in SAE. METHODS The SAE model was performed by cecal ligation and puncture (CLP), and Mdivi-1 was administrated via intraperitoneal injection. Morris water maze was performed to assess cognitive function. Mitochondrial morphology was observed by electron microscope or MitoTracker staining. The qRT-PCR, immunofluorescence staining, and western blots were used to detect the inflammatory factors and protein content, respectively. Flow cytometry was used to detect the polarization of hippocampal microglia. Bioinformatics analysis was used to verify hypotheses. RESULTS Mdivi-1 administration alleviated sepsis-induced mitochondrial fragmentation, microglia activation, polarization, and cognitive damage. The mechanisms study showed neuroinflammation and oxidative stress were suppressed via NF-κB and Keap1/Nrf2/HO-1 pathways following Mdivi-1 administration; meanwhile, pyroptosis in microglia was reduced, which was associated with enhanced autophagosome formation via p62 elevation following Mdivi-1 administration. CONCLUSION Inhibition of microglia mitochondrial fragmentation is beneficial to SAE cognitive disturbance, the mechanisms are related to alleviating neuroinflammation, oxidative stress, and pyroptosis.
Collapse
Affiliation(s)
- Chen Hong
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Li Wang
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Xiaowei Zhou
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Liyong Zou
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Xinming Xiang
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Haoyue Deng
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Qinghui Li
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Yue Wu
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Liangming Liu
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| | - Tao Li
- Shock and Transfusion Department, Research Institute of Surgery, Daping HospitalArmy Medical UniversityChongqingChina
| |
Collapse
|
6
|
Spector L, Subramanian N. Revealing the dance of NLRP3: spatiotemporal patterns in inflammasome activation. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00053. [PMID: 39816134 PMCID: PMC11731036 DOI: 10.1097/in9.0000000000000053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/01/2024] [Indexed: 01/18/2025]
Abstract
The nucleotide-binding domain, leucine-rich repeat, and pyrin domain containing-protein 3 (NLRP3) inflammasome is a multiprotein complex that plays a critical role in the innate immune response to both infections and sterile stressors. Dysregulated NLRP3 activation has been implicated in a variety of autoimmune and inflammatory diseases, including cryopyrin-associated periodic fever syndromes, diabetes, atherosclerosis, Alzheimer's disease, inflammatory bowel disease, and cancer. Consequently, fine-tuning NLRP3 activity holds significant therapeutic potential. Studies have implicated several organelles, including mitochondria, lysosomes, the endoplasmic reticulum (ER), the Golgi apparatus, endosomes, and the centrosome, in NLRP3 localization and inflammasome assembly. However, reports of conflict and many factors regulating interactions between NLRP3 and subcellular organelles remain unknown. This review synthesizes the current understanding of NLRP3 spatiotemporal dynamics, focusing on recent literature that elucidates the roles of subcellular localization and organelle stress in NLRP3 signaling and its crosstalk with other innate immune pathways converging at these organelles.
Collapse
Affiliation(s)
- Lauren Spector
- Institute for Systems Biology, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Naeha Subramanian
- Institute for Systems Biology, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| |
Collapse
|
7
|
Dong J, Tong W, Liu M, Liu M, Liu J, Jin X, Chen J, Jia H, Gao M, Wei M, Duan Y, Zhong X. Endosomal traffic disorders: a driving force behind neurodegenerative diseases. Transl Neurodegener 2024; 13:66. [PMID: 39716330 DOI: 10.1186/s40035-024-00460-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024] Open
Abstract
Endosomes are crucial sites for intracellular material sorting and transportation. Endosomal transport is a critical process involved in the selective uptake, processing, and intracellular transport of substances. The equilibrium between endocytosis and circulation mediated by the endosome-centered transport pathway plays a significant role in cell homeostasis, signal transduction, and immune response. In recent years, there have been hints linking endosomal transport abnormalities to neurodegenerative diseases, including Alzheimer's disease. Nonetheless, the related mechanisms remain unclear. Here, we provide an overview of endosomal-centered transport pathways and highlight potential physiological processes regulated by these pathways, with a particular focus on the correlation of endosomal trafficking disorders with common pathological features of neurodegenerative diseases. Additionally, we summarize potential therapeutic agents targeting endosomal trafficking for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jianru Dong
- School of Pharmacy, China Medical University, Shenyang, 110122, China
- Weifang Hospital of Traditional Chinese Medicine, Weifang, 261000, China
| | - Weiwei Tong
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, 110069, China
| | - Mingyan Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Mengyu Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Jinyue Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ju Chen
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Huachao Jia
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Menglin Gao
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Medical Diagnosis and Treatment Center, Shenyang, 110167, China.
| | - Ying Duan
- Liaoning Maternal and Child Health Hospital, Shenyang, 110005, China.
| | - Xin Zhong
- School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
8
|
Haque MF, Benjaskulluecha S, Boonmee A, Kongkavitoon P, Wongprom B, Pattarakankul T, Ongratanaphol R, Sri-Ngern-Ngam K, Pongma C, Saechue B, Kueanjinda P, Kobayashi T, Leelahavanichkul A, Palaga T. Loss of O 6-methylguanine DNA methyltransferase (MGMT) in macrophages alters responses to TLR3 stimulation and enhances DNA double-strand breaks and mitophagy. Sci Rep 2024; 14:27492. [PMID: 39528715 PMCID: PMC11554780 DOI: 10.1038/s41598-024-78885-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
O6-methylguanine-DNA methyltransferase (MGMT) is a DNA damage repair enzyme. The roles of this enzyme in immune cells remain unclear. In this study, we explored the roles of MGMT in bone marrow-derived murine macrophages (BMMs) via the use of MGMT knockout (KO) mice. Loss of MGMT altered the response to TLR3 agonists (poly (I:C)), such as dampening the production of TNFα and IL-6. Increased DNA double-strand breaks (DSBs) were observed in MGMT-KO macrophages but did not result in increased cell death. MGMT localized to both nuclei and mitochondria at increasing levels during poly (I:C) stimulation. MGMT deficiency increased the production of mitochondrial reactive oxygen species (mtROS), which was correlated with increased mitophagy. The underlying mechanism involves mediation through activation of the AMPKα pathway. Taken together, our findings reveal the roles of MGMT in macrophages in regulating the response to TLR3, which links DSBs to mtROS and mitophagy via the AMPKα pathway. These roles may have consequences for the inflammatory response and chronic inflammation.
Collapse
Affiliation(s)
- Md Fazlul Haque
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Zoology, Faculty of Biological Sciences, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Salisa Benjaskulluecha
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Atsadang Boonmee
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pornrat Kongkavitoon
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Benjawan Wongprom
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thitiporn Pattarakankul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Advanced Materials and Biointerfaces, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rahat Ongratanaphol
- Program of Bachelor of Science in Applied Chemistry (BSAC), Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kittitach Sri-Ngern-Ngam
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chitsuda Pongma
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Benjawan Saechue
- One Health Research Unit, Faculty of Veterinary Science, Mahasarakham University, Mahasarakham, 44000, Thailand
| | - Patipark Kueanjinda
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Research Center for GLOBAL and LOCAL Infectious Diseases, Oita University, Oita, 879-5593, Japan
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
9
|
Rao T, Yang W, Ma X, Jiang X, Jiang S, Xu S. Bergapten attenuates hemorrhagic shock induced multi-organ injury by inhibiting NLRP3 inflammasome activation and pyroptosis. Int Immunopharmacol 2024; 140:112839. [PMID: 39126737 DOI: 10.1016/j.intimp.2024.112839] [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: 04/09/2024] [Revised: 07/20/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVES Treatment of hemorrhagic shock (HS) induced multi-organ injury remains a challenge. Bergapten (BeG) is a bioactive coumarin-derived compound, and previous articles have suggested that BeG may serve as a prospective therapeutic modality for HS. This study was designed to investigate the efficacy of BeG in the treatment of HS and its underlying mechanisms. METHODS In this research, we established a rat model of HS, following which we assessed the protective effects of BeG on HS induced multi-organ injury. Subsequently, we scrutinized the activation of NLRP3 inflammasomes and pyroptosis in damaged organs. Additionally, we conducted examinations of AMPK and the downstream mitophagy pathway in damaged organs. Finally, we established a hypoxia/reoxygenation (H/R) model in HK-2 cells to simulate the in vitro HS process. Following AMPK inhibition with compound C, we evaluated the levels of mitophagy and cellular pyroptosis in BeG-treated HK-2 cells subjected to H/R. RESULTS BeG treatment alleviated HS induced multi-organ injury. Subsequent analyses indicated that the therapeutic effects of BeG were related to the attenuation of NLRP3 inflammasome activation and pyroptosis. Additionally, we found BeG treatment stimulated the phosphorylation of AMPK, thereby enhancing mitophagy. Lastly, we found that the inhibition of AMPK in vitro attenuates BeG's enhancement of mitophagy and its suppression of pyroptosis. CONCLUSION Our research indicates that BeG has the potential to alleviate multi-organ injury induced by HS. The protective effect of BeG is likely associated with its promotion of mitophagy through AMPK activation, thereby inhibiting NLRP3 inflammasome-mediated pyroptosis.
Collapse
Affiliation(s)
- Taiwen Rao
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Wei Yang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Ximei Ma
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Xiangkang Jiang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China
| | - Shouyin Jiang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China.
| | - Shanxiang Xu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China.
| |
Collapse
|
10
|
Wu KKL, Xu X, Wu M, Li X, Hoque M, Li GHY, Lian Q, Long K, Zhou T, Piao H, Xu A, Hui HX, Cheng KKY. MDM2 induces pro-inflammatory and glycolytic responses in M1 macrophages by integrating iNOS-nitric oxide and HIF-1α pathways in mice. Nat Commun 2024; 15:8624. [PMID: 39366973 PMCID: PMC11452520 DOI: 10.1038/s41467-024-53006-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 09/28/2024] [Indexed: 10/06/2024] Open
Abstract
M1 macrophages induce protective immunity against infection, but also contribute to metabolic and inflammatory diseases. Here we show that the E3 ubiquitin ligase, MDM2, promotes the glycolytic and inflammatory activities of M1 macrophage by increasing the production of IL-1β, MCP-1 and nitric oxide (NO). Mechanistically, MDM2 triggers the ubiquitination and degradation of E3 ligase, SPSB2, to stabilize iNOS and increases production of NO, which s-nitrosylates and activates HIF-1α for triggering the glycolytic and pro-inflammatory programs in M1 macrophages. Myeloid-specific haplodeletion of MDM2 in mice not only blunts LPS-induced endotoxemia and NO production, but also alleviates obesity-induced adipose tissue-resident macrophage inflammation. By contrast, MDM2 haplodeletion induces higher mortality, tissue damage and bacterial burden, and also suppresses M1 macrophage response, in the cecal ligation and puncture-induced sepsis mouse model. Our findings thus identify MDM2 as an activator of glycolytic and inflammatory responses in M1 macrophages by connecting the iNOS-NO and HIF-1α pathways.
Collapse
Affiliation(s)
- Kelvin Ka-Lok Wu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaofan Xu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Manyin Wu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital Fudan University, Shanghai, China
| | - Moinul Hoque
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Gloria Hoi Yee Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Qizhou Lian
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Shenzhen, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Prenatal Diagnostic Center and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Kekao Long
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Tongxi Zhou
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Hailong Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Beijing, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Hannah Xiaoyan Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kenneth King-Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China.
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
11
|
Li Z, Liang S, Ke L, Wang M, Gao K, Li D, Xu Z, Li N, Zhang P, Cheng W. Cell life-or-death events in osteoporosis: All roads lead to mitochondrial dynamics. Pharmacol Res 2024; 208:107383. [PMID: 39214266 DOI: 10.1016/j.phrs.2024.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria exhibit heterogeneous shapes and networks within and among cell types and tissues, also in normal or osteoporotic bone tissues with complex cell types. This dynamic characteristic is determined by the high plasticity provided by mitochondrial dynamics and is stemmed from responding to the survival and functional requirements of various bone cells in a specific microenvironments. In contrast, mitochondrial dysfunction, induced by dysregulation of mitochondrial dynamics, may act as a trigger of cell death signals, including common apoptosis and other forms of programmed cell death (PCD). These PCD processes consisting of tightly structured cascade gene expression events, can further influence the bone remodeling by facilitating the death of various bone cells. Mitochondrial dynamics, therefore, drive the bone cells to stand at the crossroads of life and death by integrating external signals and altering metabolism, shape, and signal-response properties of mitochondria. This implies that targeting mitochondrial dynamics displays significant potential in treatment of osteoporosis. Considerable effort has been made in osteoporosis to emphasize the parallel roles of mitochondria in regulating energy metabolism, calcium signal transduction, oxidative stress, inflammation, and cell death. However, the emerging field of mitochondrial dynamics-related PCD is not well understood. Herein, to bridge the gap, we outline the latest knowledge on mitochondrial dynamics regulating bone cell life or death during normal bone remodeling and osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050011, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000, China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300, China.
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
12
|
Jin M, Wu X, Hu J, Chen Y, Yang B, Cheng C, Yang M, Zhang X. EGFR-MEK1/2 cascade negatively regulates bactericidal function of bone marrow macrophages in mice with Staphylococcus aureus osteomyelitis. PLoS Pathog 2024; 20:e1012437. [PMID: 39102432 PMCID: PMC11326603 DOI: 10.1371/journal.ppat.1012437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/15/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024] Open
Abstract
The ability of Staphylococcus aureus (S. aureus) to survive within macrophages is a critical strategy for immune evasion, contributing to the pathogenesis and progression of osteomyelitis. However, the underlying mechanisms remain poorly characterized. This study discovered that inhibiting the MEK1/2 pathway reduced bacterial load and mitigated bone destruction in a mouse model of S. aureus osteomyelitis. Histological staining revealed increased phosphorylated MEK1/2 levels in bone marrow macrophages surrounding abscess in the mouse model of S. aureus osteomyelitis. Activation of MEK1/2 pathway and its roles in impairing macrophage bactericidal function were confirmed in primary mouse bone marrow-derived macrophages (BMDMs). Transcriptome analysis and in vitro experiments demonstrated that S. aureus activates the MEK1/2 pathway through EGFR signaling. Moreover, we found that excessive activation of EGFR-MEK1/2 cascade downregulates mitochondrial reactive oxygen species (mtROS) levels by suppressing Chek2 expression, thereby impairing macrophage bactericidal function. Furthermore, pharmacological inhibition of EGFR signaling prevented upregulation of phosphorylated MEK1/2 and restored Chek2 expression in macrophages, significantly enhancing S. aureus clearance and improving bone microstructure in vivo. These findings highlight the critical role of the EGFR-MEK1/2 cascade in host immune defense against S. aureus, suggesting that S. aureus may reduce mtROS levels by overactivating the EGFR-MEK1/2 cascade, thereby suppressing macrophage bactericidal function. Therefore, combining EGFR-MEK1/2 pathway blockade with antibiotics could represent an effective therapeutic approach for the treatment of S. aureus osteomyelitis.
Collapse
Affiliation(s)
- Mingchao Jin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaohu Wu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Hu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yijie Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingsheng Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chubin Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mankai Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianrong Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Xiong H, Li X, Mou X, Huang C, Yi S, Xiong X, Zhou Y, Chen Y. Syringic acid suppresses Cutibacterium acnes-induced inflammation in human keratinocytes via regulating the NLRP3/caspase-1/IL-1β signaling axis by activating PPARγ/Nrf2-antioxidant pathway. Int Immunopharmacol 2024; 139:112708. [PMID: 39033661 DOI: 10.1016/j.intimp.2024.112708] [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: 01/22/2024] [Revised: 06/12/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Our previous studies have demonstrated a strong relationship betweenCutibacterium acnes(C. acnes), oxidative stress, and acne inflammation. Syringic acid (SA) is a plant widely used for its antimicrobial, anti-inflammatory, and antioxidant activities, but lacking data on acne. This study aims to investigate the effect and mechanism of SA on acne inflammation induced by C. acnes in vitro and in vivo. METHODS After using the SA to expose HaCaT keratinocytes, we reevaluated the effect of the SA on cell viability, cell apoptosis, ROS, CAT, SOD, and other inflammatory variables in the heat-killed C. acnes-treated HaCaT cells. Next, to induce mice with acne inflammation, ICR mice were given an intradermal injection of live C. acnes into their right ears. The effect of SA on this inflammation was then examined. Moreover, we explored the mechanism of SA on PPARγ/Nrf2 and NLRP3/caspase-1/IL-1β pathways by ELISA, immunofluorescence microscopy, and western blot assay. RESULTS Heat-killed C. acnes triggered remarkable cell apoptosis, ROS production, interleukin (IL)-1β, IL-18, IL-6, and TNF-α release, reduced SOD and CAT activity, and upregulated the expression of proteins in HaCaT cells, including up-regulating IL-1β, PPARγ, Nrf2, HO-1, NQO1, NLRP3, and caspase-1, whereas SA inhibited these effects by partially impairing PPARγ activation. In addition, PPARγ silencing decreased C. acnes-induced IL-1β secretion and the production of intracellular ROS, down-regulating the expression of Nrf2. Nrf2 activator (SFN) enhanced anti-inflammatory activity through antioxidant mechanisms, boosting intracellular ROS production, reducing SOD and CAT activity, and promoting the increase in ROS, HO-1, NQO1, and IL-1β levels, while PPARγ inhibitor (GW662) effectively inhibited this effect in heat-killed C. acnes-treated cells. Finally, SA also exhibited notable improvements in ear redness, swelling, and the expression of PPARγ, NLRP3, and IL-1β in vivo. CONCLUSIONS SA inhibited C. acnes-induced inflammation via regulating the NLRP3/caspase-1/IL-1β signaling axis by activating the PPARγ/Nrf2-antioxidant pathway, suggesting a new treatment possibility for acne vulgaris.
Collapse
Affiliation(s)
- Haojun Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Xingzhu Mou
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Cancan Huang
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Sha Yi
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yingshun Zhou
- Department of Pathogenic Biology, School of Basic Medicine and Public Center of Experimental Technology of Pathogen Biology technology platform, Southwest Medical University, Luzhou 646000, China.
| | - Yan Chen
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
14
|
Li T, Qu J, Hu C, Pang J, Qian Y, Li Y, Peng Z. Macrophage migration inhibitory factor (MIF) suppresses mitophagy through disturbing the protein interaction of PINK1-Parkin in sepsis-associated acute kidney injury. Cell Death Dis 2024; 15:473. [PMID: 38956064 PMCID: PMC11220046 DOI: 10.1038/s41419-024-06826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/04/2024]
Abstract
Damage to renal tubular epithelial cells (RTECs) signaled the onset and progression of sepsis-associated acute kidney injury (SA-AKI). Recent research on mitochondria has revealed that mitophagy plays a crucial physiological role in alleviating injury to RTECs and it is suppressed progressively by the inflammation response in SA-AKI. However, the mechanism by which inflammation influences mitophagy remains poorly understood. We examined how macrophage migration inhibitory factor (MIF), a pro-inflammatory protein, influences the PINK1-Parkin pathway of mitophagy by studying protein-protein interactions when MIF was inhibited or overexpressed. Surprisingly, elevated levels of MIF were found to directly bind to PINK1, disrupting its interaction with Parkin. This interference hindered the recruitment of Parkin to mitochondria and impeded the initiation of mitophagy. Furthermore, this outcome led to significant apoptosis of RTECs, which could, however, be reversed by an MIF inhibitor ISO-1 and/or a new mitophagy activator T0467. These findings highlight the detrimental impact of MIF on renal damage through its disruption of the interaction between PINK1 and Parkin, and the therapeutic potential of ISO-1 and T0467 in mitigating SA-AKI. This study offers a fresh perspective on treating SA-AKI by targeting MIF and mitophagy.
Collapse
Affiliation(s)
- Tianlong Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, 430071, China
| | - Jiachen Qu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, 430071, China
| | - Chang Hu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, 430071, China
| | - Jingjing Pang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, 430071, China
| | - Yaoyao Qian
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, 430071, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, 430071, China.
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei province, 430071, China.
| |
Collapse
|
15
|
Ramachandran R, Manan A, Kim J, Choi S. NLRP3 inflammasome: a key player in the pathogenesis of life-style disorders. Exp Mol Med 2024; 56:1488-1500. [PMID: 38945951 PMCID: PMC11297159 DOI: 10.1038/s12276-024-01261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 07/02/2024] Open
Abstract
Proinflammatory cytokines and chemokines play a crucial role in regulating the inflammatory response, which is essential for the proper functioning of our immune system. When infections or threats to the body's defense mechanisms are detected, the innate immune system takes the lead. However, an excessive inflammatory response can lead to the production of high concentrations of cytotoxic molecules, resulting in tissue damage. Inflammasomes are significant contributors to innate immunity, and one of the most extensively studied inflammasome complexes is NOD-like receptor 3 (NLRP3). NLRP3 has a wide range of recognition mechanisms that streamline immune activation and eliminate pathogens. These cytosolic multiprotein complexes are composed of effector, adaptor, and sensor proteins, which are crucial for identifying intracellular bacterial breakdown products and initiating an innate immune cascade. To understand the diverse behavior of NLRP3 activation and its significance in the development of lifestyle-related diseases, one must delve into the study of the immune response and apoptosis mediated by the release of proinflammatory cytokines. In this review, we briefly explore the immune response in the context of lifestyle associated disorders such as obesity, hyperlipidemia, diabetes, chronic respiratory disease, oral disease, and cardiovascular disease.
Collapse
Affiliation(s)
- Rajath Ramachandran
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea.
| | - Abdul Manan
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Jei Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon, 16502, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea.
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon, 16502, Korea.
| |
Collapse
|
16
|
Kraft BD. "Mitolocalization": A Novel Marker of Mitochondrial Quality Control? CHEST CRITICAL CARE 2024; 2:100073. [PMID: 39301036 PMCID: PMC11412265 DOI: 10.1016/j.chstcc.2024.100073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Affiliation(s)
- Bryan D Kraft
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine
| |
Collapse
|
17
|
Lv T, Fan X, He C, Zhu S, Xiong X, Yan W, Liu M, Xu H, Shi R, He Q. SLC7A11-ROS/αKG-AMPK axis regulates liver inflammation through mitophagy and impairs liver fibrosis and NASH progression. Redox Biol 2024; 72:103159. [PMID: 38642501 PMCID: PMC11047786 DOI: 10.1016/j.redox.2024.103159] [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: 03/25/2024] [Revised: 04/10/2024] [Accepted: 04/13/2024] [Indexed: 04/22/2024] Open
Abstract
The changes of inflammation and metabolism are two features in nonalcoholic steatohepatitis (NASH). However, how they interact to regulate NASH progression remains largely unknown. Our works have demonstrated the importance of solute carrier family 7 member 11 (SLC7A11) in inflammation and metabolism. Nevertheless, whether SLC7A11 regulates NASH progression through mediating inflammation and metabolism is unclear. In this study, we found that SLC7A11 expression was increased in liver samples from patients with NASH. Upregulated SLC7A11 level was also detected in two murine NASH models. Functional studies showed that SLC7A11 knockdown or knockout had augmented steatohepatitis with suppression of inflammatory markers in mice. However, overexpression of SLC7A11 dramatically alleviated diet-induced NASH pathogenesis. Mechanically, SLC7A11 decreased reactive oxygen species (ROS) level and promoted α-ketoglutarate (αKG)/prolyl hydroxylase (PHD) activity, which activated AMPK pathway. Furthermore, SLC7A11 impaired expression of NLRP3 inflammasome components through AMPK-mitophagy axis. IL-1β release through NLRP3 inflammasome recruited myeloid cells and promoted hepatic stellate cells (HSCs) activation, which contributed to the progression of liver injury and fibrosis. Anti-IL-1β and anakinra might attenuate the hepatic inflammatory response evoked by SLC7A11 knockdown. Moreover, the upregulation of SLC7A11 in NASH was contributed by lipid overload-induced JNK-c-Jun pathway. In conclusions, SLC7A11 acts as a protective factor in controlling the development of NASH. Upregulation of SLC7A11 is protective by regulating oxidation, αKG and energy metabolism, decreasing inflammation and fibrosis.
Collapse
Affiliation(s)
- Tingting Lv
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Department of Cancer Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Department of Gastroenterology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xiude Fan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Chang He
- Medical College, Nantong University, Nantong, Jiangsu, 226001, China
| | - Suwei Zhu
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xiaofeng Xiong
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wei Yan
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Mei Liu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Hongwei Xu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| | - Ruihua Shi
- Department of Gastroenterology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, 210009, China.
| | - Qin He
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Department of Gastroenterology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
18
|
Zimmermann A, Madeo F, Diwan A, Sadoshima J, Sedej S, Kroemer G, Abdellatif M. Metabolic control of mitophagy. Eur J Clin Invest 2024; 54:e14138. [PMID: 38041247 DOI: 10.1111/eci.14138] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023]
Abstract
Mitochondrial dysfunction is a major hallmark of ageing and related chronic disorders. Controlled removal of damaged mitochondria by the autophagic machinery, a process known as mitophagy, is vital for mitochondrial homeostasis and cell survival. The central role of mitochondria in cellular metabolism places mitochondrial removal at the interface of key metabolic pathways affecting the biosynthesis or catabolism of acetyl-coenzyme A, nicotinamide adenine dinucleotide, polyamines, as well as fatty acids and amino acids. Molecular switches that integrate the metabolic status of the cell, like AMP-dependent protein kinase, protein kinase A, mechanistic target of rapamycin and sirtuins, have also emerged as important regulators of mitophagy. In this review, we discuss how metabolic regulation intersects with mitophagy. We place special emphasis on the metabolic regulatory circuits that may be therapeutically targeted to delay ageing and mitochondria-associated chronic diseases. Moreover, we identify outstanding knowledge gaps, such as the ill-defined distinction between basal and damage-induced mitophagy, which must be resolved to boost progress in this area.
Collapse
Affiliation(s)
- Andreas Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth-University of Graz, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth-University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Abhinav Diwan
- Division of Cardiology and Center for Cardiovascular Research, Washington University School of Medicine, and John Cochran Veterans Affairs Medical Center, St. Louis, Missouri, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Simon Sedej
- BioTechMed Graz, Graz, Austria
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Department of Biology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, Paris, France
| | - Mahmoud Abdellatif
- BioTechMed Graz, Graz, Austria
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
| |
Collapse
|
19
|
Zhong H, Ji J, Zhuang J, Xiong Z, Xie P, Liu X, Zheng J, Tian W, Hong X, Tang J. Tissue-resident macrophages exacerbate lung injury after remote sterile damage. Cell Mol Immunol 2024; 21:332-348. [PMID: 38228746 PMCID: PMC10979030 DOI: 10.1038/s41423-024-01125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/26/2023] [Indexed: 01/18/2024] Open
Abstract
Remote organ injury, which is a common secondary complication of sterile tissue damage, is a major cause of poor prognosis and is difficult to manage. Here, we report the critical role of tissue-resident macrophages in lung injury after trauma or stroke through the inflammatory response. We found that depleting tissue-resident macrophages rather than disrupting the recruitment of monocyte-derived macrophages attenuated lung injury after trauma or stroke. Our findings revealed that the release of circulating alarmins from sites of distant sterile tissue damage triggered an inflammatory response in lung-resident macrophages by binding to receptor for advanced glycation end products (RAGE) on the membrane, which activated epidermal growth factor receptor (EGFR). Mechanistically, ligand-activated RAGE triggered EGFR activation through an interaction, leading to Rab5-mediated RAGE internalization and EGFR phosphorylation, which subsequently recruited and activated P38; this, in turn, promoted RAGE translation and trafficking to the plasma membrane to increase the cellular response to RAGE ligands, consequently exacerbating inflammation. Our study also showed that the loss of RAGE or EGFR expression by adoptive transfer of macrophages, blocking the function of RAGE with a neutralizing antibody, or pharmacological inhibition of EGFR activation in macrophages could protect against trauma- or stroke-induced remote lung injury. Therefore, our study revealed that targeting the RAGE-EGFR signaling pathway in tissue-resident macrophages is a potential therapeutic approach for treating secondary complications of sterile damage.
Collapse
Affiliation(s)
- Hanhui Zhong
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jingjing Ji
- The Department of Critical Care Medicine, General Hospital of Southern Theater Command of PLA, Guangzhou, China
| | - Jinling Zhuang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Ziying Xiong
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Pengyun Xie
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiaolei Liu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jundi Zheng
- The Department of Respiratory Medicine, Guangdong Provincial Hospital of Integrated Chinese and Western Medicine, Foshan, China
| | - Wangli Tian
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiaoyang Hong
- Pediatric Intensive Care Unit, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China.
| | - Jing Tang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
20
|
Ma L, Han T, Zhan YA. Mechanism and role of mitophagy in the development of severe infection. Cell Death Discov 2024; 10:88. [PMID: 38374038 PMCID: PMC10876966 DOI: 10.1038/s41420-024-01844-4] [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: 10/23/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/21/2024] Open
Abstract
Mitochondria produce adenosine triphosphate and potentially contribute to proinflammatory responses and cell death. Mitophagy, as a conservative phenomenon, scavenges waste mitochondria and their components in the cell. Recent studies suggest that severe infections develop alongside mitochondrial dysfunction and mitophagy abnormalities. Restoring mitophagy protects against excessive inflammation and multiple organ failure in sepsis. Here, we review the normal mitophagy process, its interaction with invading microorganisms and the immune system, and summarize the mechanism of mitophagy dysfunction during severe infection. We highlight critical role of normal mitophagy in preventing severe infection.
Collapse
Affiliation(s)
- Lixiu Ma
- Department of Respiratory and Critical Care Medicine, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Tianyu Han
- Jiangxi Institute of Respiratory Disease, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yi-An Zhan
- Department of Respiratory and Critical Care Medicine, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
21
|
Shi P, Tian Y, Xu F, Liu LN, Wu WH, Shi YZ, Dai AQ, Fang HY, Li KX, Xu C. Assessment of pathogenicity and functional characterization of APPL1 gene mutations in diabetic patients. World J Diabetes 2024; 15:275-286. [PMID: 38464380 PMCID: PMC10921161 DOI: 10.4239/wjd.v15.i2.275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/22/2023] [Accepted: 01/09/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1) plays a crucial role in regulating insulin signaling and glucose metabolism. Mutations in the APPL1 gene have been associated with the development of maturity-onset diabetes of the young type 14 (MODY14). Currently, only two mutations [c.1655T>A (p.Leu552*) and c.281G>A p.(Asp94Asn)] have been identified in association with this disease. Given the limited understanding of MODY14, it is imperative to identify additional cases and carry out comprehensive research on MODY14 and APPL1 mutations. AIM To assess the pathogenicity of APPL1 gene mutations in diabetic patients and to characterize the functional role of the APPL1 domain. METHODS Patients exhibiting clinical signs and a medical history suggestive of MODY were screened for the study. Whole exome sequencing was performed on the patients as well as their family members. The pathogenicity of the identified APPL1 variants was predicted on the basis of bioinformatics analysis. In addition, the pathogenicity of the novel APPL1 variant was preliminarily evaluated through in vitro functional experiments. Finally, the impact of these variants on APPL1 protein expression and the insulin pathway were assessed, and the potential mechanism underlying the interaction between the APPL1 protein and the insulin receptor was further explored. RESULTS A total of five novel mutations were identified, including four missense mutations (Asp632Tyr, Arg633His, Arg532Gln, and Ile642Met) and one intronic mutation (1153-16A>T). Pathogenicity prediction analysis revealed that the Arg532Gln was pathogenic across all predictions. The Asp632Tyr and Arg633His variants also had pathogenicity based on MutationTaster. In addition, multiple alignment of amino acid sequences showed that the Arg532Gln, Asp632Tyr, and Arg633His variants were conserved across different species. Moreover, in in vitro functional experiments, both the c.1894G>T (at Asp632Tyr) and c.1595G>A (at Arg532Gln) mutations were found to downregulate the expression of APPL1 on both protein and mRNA levels, indicating their pathogenic nature. Therefore, based on the patient's clinical and family history, combined with the results from bioinformatics analysis and functional experiment, the c.1894G>T (at Asp632Tyr) and c.1595G>A (at Arg532Gln) mutations were classified as pathogenic mutations. Importantly, all these mutations were located within the phosphotyrosine-binding domain of APPL1, which plays a critical role in the insulin sensitization effect. CONCLUSION This study provided new insights into the pathogenicity of APPL1 gene mutations in diabetes and revealed a potential target for the diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Ping Shi
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Yang Tian
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Feng Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Lu-Na Liu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Wan-Hong Wu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Ying-Zhou Shi
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - An-Qi Dai
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Hang-Yu Fang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Kun-Xia Li
- Department of Pediatric, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264099, Shandong Province, China
| | - Chao Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| |
Collapse
|
22
|
Bornstein B, Watkins B, Passini FS, Blecher R, Assaraf E, Sui XM, Brumfeld V, Tsoory M, Kröger S, Zelzer E. The mechanosensitive ion channel ASIC2 mediates both proprioceptive sensing and spinal alignment. Exp Physiol 2024; 109:135-147. [PMID: 36951012 PMCID: PMC10988735 DOI: 10.1113/ep090776] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/20/2023] [Indexed: 03/24/2023]
Abstract
By translating mechanical forces into molecular signals, proprioceptive neurons provide the CNS with information on muscle length and tension, which is necessary to control posture and movement. However, the identities of the molecular players that mediate proprioceptive sensing are largely unknown. Here, we confirm the expression of the mechanosensitive ion channel ASIC2 in proprioceptive sensory neurons. By combining in vivo proprioception-related functional tests with ex vivo electrophysiological analyses of muscle spindles, we showed that mice lacking Asic2 display impairments in muscle spindle responses to stretch and motor coordination tasks. Finally, analysis of skeletons of Asic2 loss-of-function mice revealed a specific effect on spinal alignment. Overall, we identify ASIC2 as a key component in proprioceptive sensing and a regulator of spine alignment.
Collapse
Affiliation(s)
- Bavat Bornstein
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| | - Bridgette Watkins
- Department of Physiological Genomics, Biomedical CenterLudwig‐Maximilians‐UniversityPlanegg‐MartinsriedGermany
| | - Fabian S. Passini
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| | - Ronen Blecher
- Orthopedic DepartmentAssuta Ashdod University Hospital, Ashdod, Israel, affiliated to Ben Gurion University of the NegevBeer ShebaIsrael
| | - Eran Assaraf
- Department of Orthopedic SurgeryShamir Medical Center, Assaf HaRofeh Campus, Zeffifin, Israel, affiliated to Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Xiao Meng Sui
- Department of Chemical Research SupportWeizmann Institute of ScienceRehovotIsrael
| | - Vlad Brumfeld
- Department of Chemical Research SupportWeizmann Institute of ScienceRehovotIsrael
| | - Michael Tsoory
- Department of Veterinary ResourcesWeizmann Institute of ScienceRehovotIsrael
| | - Stephan Kröger
- Department of Physiological Genomics, Biomedical CenterLudwig‐Maximilians‐UniversityPlanegg‐MartinsriedGermany
| | - Elazar Zelzer
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
23
|
Kuang X, Chen S, Ye Q. The Role of Histone Deacetylases in NLRP3 Inflammasomesmediated Epilepsy. Curr Mol Med 2024; 24:980-1003. [PMID: 37519210 DOI: 10.2174/1566524023666230731095431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023]
Abstract
Epilepsy is one of the most common brain disorders that not only causes death worldwide, but also affects the daily lives of patients. Previous studies have revealed that inflammation plays an important role in the pathophysiology of epilepsy. Activation of inflammasomes can promote neuroinflammation by boosting the maturation of caspase-1 and the secretion of various inflammatory effectors, including chemokines, interleukins, and tumor necrosis factors. With the in-depth research on the mechanism of inflammasomes in the development of epilepsy, it has been discovered that NLRP3 inflammasomes may induce epilepsy by mediating neuronal inflammatory injury, neuronal loss and blood-brain barrier dysfunction. Therefore, blocking the activation of the NLRP3 inflammasomes may be a new epilepsy treatment strategy. However, the drugs that specifically block NLRP3 inflammasomes assembly has not been approved for clinical use. In this review, the mechanism of how HDACs, an inflammatory regulator, regulates the activation of NLRP3 inflammasome is summarized. It helps to explore the mechanism of the HDAC inhibitors inhibiting brain inflammatory damage so as to provide a potential therapeutic strategy for controlling the development of epilepsy.
Collapse
Affiliation(s)
- Xi Kuang
- Hainan Health Vocational College,Haikou, Hainan, 570311, China
| | - Shuang Chen
- Hubei Provincial Hospital of Integrated Chinese and Western Medicine, 430022, Hubei, China
| | - Qingmei Ye
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| |
Collapse
|
24
|
She M, Huang M, Zhang J, Yan Y, Zhou L, Zhang M, Yang Y, Wang D. Astragulus embranaceus (Fisch.) Bge-Dioscorea opposita Thunb herb pair ameliorates sarcopenia in senile type 2 diabetes mellitus through Rab5a/mTOR-mediated mitochondrial dysfunction. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116737. [PMID: 37295571 DOI: 10.1016/j.jep.2023.116737] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/15/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The combination of Astragulus embranaceus (Fisch.) Bge (Huangqi) and Dioscorea opposita Thunb (Shanyao) are one of the most widely accepted herb pairs in traditional Chinese medicine prescriptions for treating sarcopenia. However, the mechanisms underlying the combination of these herbs for anti-sarcopenia treatment are not yet fully understood. AIM OF THE STUDY To investigate the potential effect of the Astragulus embranaceus (Fisch.) Bge and Dioscorea opposita Thunb herb pair (Ast-Dio) on sarcopenia in mice that have been induced with senile type 2 diabetes mellitus, as well as to explore the underlying mechanisms related to the Rab5a/mTOR signaling pathway and mitochondrial quality control. MATERIALS AND METHODS Network pharmacology was utilized to identify the main active ingredients of Ast-Dio and potential therapeutic targets for sarcopenia. Gene Ontology function and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were conducted to explore the underlying mechanisms of Ast-Dio in treating sarcopenia. The high-performance liquid chromatography method coupled with triple-quadrupole tandem mass spectrometry was developed to quantify the major constituents of Ast-Dio. Male C57/BL6 mice, aged 12 months, induced with type 2 diabetes mellitus via streptozotocin were divided into three groups for 8 weeks: the model group, Ast-Dio treatment group (7.8 g/kg), and metformin treatment group (100 mg/kg). Normal control groups included mice aged 3 and 12 months, respectively. The study monitored changes in fasting blood glucose levels, grip strength, and body weight during 8 weeks of intragastric administration. Liver and kidney function in mice was evaluated by measuring the levels of serum creatinine, alanine transaminase, and aspartate transaminase. Skeletal muscle mass condition was evaluated by muscle weight, and hematoxylin and eosin staining. Protein and mRNA expressions related to muscle atrophy, mitochondrial quality control, and the Rab5a/mTOR signaling pathway were detected using immunofluorescence staining, immunohistochemical staining, Western blotting, and quantitative real-time polymerase chain reaction. In addition, transmission electron microscopy was employed to investigate the condition of mitochondria in the groups. RESULTS Through the prediction analysis of network pharmacology, we identified mTOR as one of the primary targets for Ast-Dio therapy of sarcopenia. Gene Ontology functional enrichment analysis revealed that mitochondrial control quality is crucial in the treatment of sarcopenia with Ast-Dio. Our findings showed that senile type 2 diabetes mellitus induced muscle mass loss and a reduction in grip strength, both of which were dramatically restored by Ast-Dio treatment. Notably, Ast-Dio increased Myogenin expression while decreasing Atrogin-1 and MuRF-1 expression. Additionally, Ast-Dio activated Rab5a/mTOR and its downstream effector AMPK. Moreover, Ast-Dio modulated mitochondrial quality control by decreasing Mitofusin-2 expression while increasing the expression of TFAM, PGC-1α, and MFF. CONCLUSIONS Our results suggest that Ast-Dio treatment may alleviate sarcopenia in mice with senile type 2 diabetes mellitus through its effects on the Rab5a/mTOR pathway and mitochondrial quality control.
Collapse
Affiliation(s)
- Meiling She
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, No. 1333, Xinhu Road, Bao 'an District, Shenzhen, Guangdong, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510000, China
| | - Minna Huang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, No. 1333, Xinhu Road, Bao 'an District, Shenzhen, Guangdong, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510000, China
| | - Jing Zhang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, No. 1333, Xinhu Road, Bao 'an District, Shenzhen, Guangdong, 518000, China
| | - Yan Yan
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, No. 1333, Xinhu Road, Bao 'an District, Shenzhen, Guangdong, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510000, China
| | - Lingli Zhou
- The First Clinical Medical College, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510000, China
| | - Meng Zhang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, No. 1333, Xinhu Road, Bao 'an District, Shenzhen, Guangdong, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510000, China.
| | - Yajun Yang
- Department of Pharmacology, Guangdong Key Laboratory for R&D of Natural Drug, Guangdong Medical University, No. 2, Wenming East Road, Xiashan District, Zhanjiang City, 524000, China.
| | - Dongtao Wang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, No. 1333, Xinhu Road, Bao 'an District, Shenzhen, Guangdong, 518000, China; School of Traditional Chinese Medicine, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou, Guangdong, 510000, China.
| |
Collapse
|
25
|
Zhang W, Jiang H, Wu G, Huang P, Wang H, An H, Liu S, Zhang W. The pathogenesis and potential therapeutic targets in sepsis. MedComm (Beijing) 2023; 4:e418. [PMID: 38020710 PMCID: PMC10661353 DOI: 10.1002/mco2.418] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as "a life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection." At present, sepsis continues to pose a grave healthcare concern worldwide. Despite the use of supportive measures in treating traditional sepsis, such as intravenous fluids, vasoactive substances, and oxygen plus antibiotics to eradicate harmful pathogens, there is an ongoing increase in both the morbidity and mortality associated with sepsis during clinical interventions. Therefore, it is urgent to design specific pharmacologic agents for the treatment of sepsis and convert them into a novel targeted treatment strategy. Herein, we provide an overview of the molecular mechanisms that may be involved in sepsis, such as the inflammatory response, immune dysfunction, complement deactivation, mitochondrial damage, and endoplasmic reticulum stress. Additionally, we highlight important targets involved in sepsis-related regulatory mechanisms, including GSDMD, HMGB1, STING, and SQSTM1, among others. We summarize the latest advancements in potential therapeutic drugs that specifically target these signaling pathways and paramount targets, covering both preclinical studies and clinical trials. In addition, this review provides a detailed description of the crosstalk and function between signaling pathways and vital targets, which provides more opportunities for the clinical development of new treatments for sepsis.
Collapse
Affiliation(s)
- Wendan Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Honghong Jiang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Gaosong Wu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Pengli Huang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Haonan Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huazhasng An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghaiChina
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosecurityShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
26
|
Cui H, Banerjee S, Xie N, Dey T, Liu RM, Sanders YY, Liu G. MafB regulates NLRP3 inflammasome activation by sustaining p62 expression in macrophages. Commun Biol 2023; 6:1047. [PMID: 37845329 PMCID: PMC10579372 DOI: 10.1038/s42003-023-05426-5] [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: 05/05/2023] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
Activation of the NLRP3 inflammasome is a two-step process: the priming and the activating. The priming step involves the induction of NLRP3 and pro-IL-1β, while the activating step leads to the full inflammasome activation triggered by a NLRP3 activator. Although mechanisms underlying the NLRP3 inflammasome activation have been increasingly clear, the regulation of this process remains incompletely understood. In this study, we find that LPS and Pseudomonas aeruginosa cause a rapid downregulation in MafB transcription in macrophages, which leads to a quick decline in the level of MafB protein because MafB is short-lived and constantly degraded by the ubiquitin/proteasome system. We find that MafB knockdown or knockout markedly enhances the NLRP3, but not the NLRP1, NLRC4, or AIM2, inflammasome activation in macrophages. Conversely, pharmacological induction of MafB diminishes the NLRP3 inflammasome activation. Mechanistically, we find that MafB sustains the expression of p62, a key mediator of autophagy/mitophagy. We find that MafB inhibits mitochondrial damage, and mitochondrial ROS production and DNA cytoplasmic release. Furthermore, we find that myeloid MafB deficient mice demonstrate increased systemic and lung IL-1β production in response to LPS treatment and P. aeruginosa infection and deficient lung P. aeruginosa clearance in vivo. In conclusion, our study demonstrates that MafB is an important negative regulator of the NLRP3 inflammasome. Our findings suggest that strategies elevating MafB may be effective to treat immune disorders due to excessive activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sami Banerjee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Na Xie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Tapan Dey
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Yan Y Sanders
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23501, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
27
|
Rosa CP, Belo TCA, Santos NCDM, Silva EN, Gasparotto J, Corsetti PP, de Almeida LA. Reactive oxygen species trigger inflammasome activation after intracellular microbial interaction. Life Sci 2023; 331:122076. [PMID: 37683723 DOI: 10.1016/j.lfs.2023.122076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/16/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
The intracellular production of reactive oxygen species (ROS), composed of oxygen-reduced molecules, is important not only because of their lethal effects on microorganisms but also due to their potential inflammatory and metabolic regulation properties. The ROS pro-inflammatory properties are associated with the second signal to inflammasome activation, leading to cleaving pro-IL-1β and pro-IL18 before their secretion, as well as gasdermin-D, leading to pyroptosis. Some microorganisms can modulate NLRP3 and AIM-2 inflammasomes through ROS production: whilst Mycobacterium bovis, Mycobacterium kansasii, Francisella novicida, Brucella abortus, Listeria monocytogenes, Influenza virus, Syncytial respiratory virus, Porcine reproductive and respiratory syndrome virus, SARS-CoV, Mayaro virus, Leishmania amazonensis and Plasmodium sp. enhance inflammasome assembly, Hepatitis B virus, Mycobacterium marinum, Mycobacterium tuberculosis, Francisella tularensis and Leishmania sp. disrupt it. This process represents a recent cornerstone in our knowledge of the immunology of intracellular pathogens, which is reviewed in this mini-review.
Collapse
Affiliation(s)
- Caio Pupin Rosa
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas 37130-001, Minas Gerais, Brazil
| | - Thiago Caetano Andrade Belo
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas 37130-001, Minas Gerais, Brazil
| | - Natália Cristina de Melo Santos
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas 37130-001, Minas Gerais, Brazil
| | - Evandro Neves Silva
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas 37130-001, Minas Gerais, Brazil
| | - Juciano Gasparotto
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas 37130-001, Minas Gerais, Brazil
| | - Patrícia Paiva Corsetti
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas 37130-001, Minas Gerais, Brazil
| | - Leonardo Augusto de Almeida
- Laboratory of Molecular Biology of Microorganisms, Federal University of Alfenas (UNIFAL), Alfenas 37130-001, Minas Gerais, Brazil.
| |
Collapse
|
28
|
Sun Y, Liu C. Application and value of hydrogen sulfide modulated autophagy in sepsis. Int Immunopharmacol 2023; 122:110662. [PMID: 37473711 DOI: 10.1016/j.intimp.2023.110662] [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: 01/26/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Sepsis is is anabnormalhost immune responsecausedbyinfection. Antibiotics, anti-viral drugs, and vasoactive drugs have always been used in the traditional treatment of sepsis, but there are no specific and effective drugs in clinical practice. Autophagy is a highly conservative process in biological evolution, and plays an important role in maintaining intracellular homeostasis and cellular self-renewal. Autophagy can remove and degrade misfolding proteins and damaged organelles in cells, providing materials for cell repair and self-renewal. Hydrogen sulfide (H2S) is a colorless gas that smells likerotteneggs. It is the third endogenous gas signal molecule discovered after nitric oxide and carbon monoxide and has become a research hotspot in recent years. H2S has a variety of biological functions and plays an important role in various physiological and pathological processes. Thereisgrowingevidencethat H2S can regulate autophagy. The intervention of autophagy is a promising therapeutic strategy to improve sepsis organ damage. This article reviews the organ protection of autophagy in sepsis and the role of H2S in regulating autophagy in sepsis, revealing that H2S intervention with autophagy may be a a worthy target in sepsis treatment.
Collapse
Affiliation(s)
- Yao Sun
- Department of Critical Care Medicine, Peking University People's Hospital, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
29
|
Luo T, Jia X, Feng WD, Wang JY, Xie F, Kong LD, Wang XJ, Lian R, Liu X, Chu YJ, Wang Y, Xu AL. Bergapten inhibits NLRP3 inflammasome activation and pyroptosis via promoting mitophagy. Acta Pharmacol Sin 2023; 44:1867-1878. [PMID: 37142684 PMCID: PMC10462717 DOI: 10.1038/s41401-023-01094-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/17/2023] [Indexed: 05/06/2023]
Abstract
Inhibition of NLRP3 inflammasome activation produces potent therapeutic effects in a wide array of inflammatory diseases. Bergapten (BeG), a furocoumarin phytohormone present in many herbal medicines and fruits, exibits anti-inflammatory activity. In this study we characterized the therapeutic potential of BeG against bacterial infection and inflammation-related disorders, and elucidated the underlying mechanisms. We showed that pre-treatment with BeG (20 μM) effectively inhibited NLRP3 inflammasome activation in both lipopolysaccharides (LPS)-primed J774A.1 cells and bone marrow-derived macrophages (BMDMs), evidenced by attenuated cleaved caspase-1 and mature IL-1β release, as well as reduced ASC speck formation and subsequent gasdermin D (GSDMD)-mediated pyroptosis. Transcriptome analysis revealed that BeG regulated the expression of genes involved in mitochondrial and reactive oxygen species (ROS) metabolism in BMDMs. Moreover, BeG treatment reversed the diminished mitochondrial activity and ROS production after NLRP3 activation, and elevated the expression of LC3-II and enhanced the co-localization of LC3 with mitochondria. Treatment with 3-methyladenine (3-MA, 5 mM) reversed the inhibitory effects of BeG on IL-1β, cleaved caspase-1 and LDH release, GSDMD-N formation as well as ROS production. In mouse model of Escherichia coli-induced sepsis and mouse model of Citrobacter rodentium-induced intestinal inflammation, pre-treatment with BeG (50 mg/kg) significantly ameliorated tissue inflammation and injury. In conclusion, BeG inhibits NLRP3 inflammasome activation and pyroptosis by promoting mitophagy and maintaining mitochondrial homeostasis. These results suggest BeG as a promising drug candidate for the treatment of bacterial infection and inflammation-related disorders.
Collapse
Affiliation(s)
- Tong Luo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin Jia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wan-di Feng
- Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jin-Yong Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fang Xie
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ling-Dong Kong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue-Jiao Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Rui Lian
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xia Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ying-Jie Chu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yao Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - An-Long Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
30
|
Abstract
According to the endosymbiotic theory, most of the DNA of the original bacterial endosymbiont has been lost or transferred to the nucleus, leaving a much smaller (∼16 kb in mammals), circular molecule that is the present-day mitochondrial DNA (mtDNA). The ability of mtDNA to escape mitochondria and integrate into the nuclear genome was discovered in budding yeast, along with genes that regulate this process. Mitochondria have emerged as key regulators of innate immunity, and it is now recognized that mtDNA released into the cytoplasm, outside of the cell, or into circulation activates multiple innate immune signaling pathways. Here, we first review the mechanisms through which mtDNA is released into the cytoplasm, including several inducible mitochondrial pores and defective mitophagy or autophagy. Next, we cover how the different forms of released mtDNA activate specific innate immune nucleic acid sensors and inflammasomes. Finally, we discuss how intracellular and extracellular mtDNA release, including circulating cell-free mtDNA that promotes systemic inflammation, are implicated in human diseases, bacterial and viral infections, senescence and aging.
Collapse
Affiliation(s)
- Laura E Newman
- Salk Institute for Biological Studies, La Jolla, California, USA;
| | - Gerald S Shadel
- Salk Institute for Biological Studies, La Jolla, California, USA;
| |
Collapse
|
31
|
Logan JM, Hopkins AM, Martini C, Sorvina A, Tewari P, Prabhakaran S, Huzzell C, Johnson IRD, Hickey SM, Ung BSY, Lazniewska J, Brooks RD, Moore CR, Caruso MC, Karageorgos L, Martin CM, O'Toole S, Bogue Edgerton L, Ward MP, Bates M, Selemidis S, Esterman A, Heffernan S, Keegan H, Ní Mhaolcatha S, O'Connor R, Malone V, Carter M, Ryan K, Clarke A, Brady N, Klebe S, Samaratunga H, Delahunt B, Sorich MJ, Moretti K, Butler LM, O'Leary JJ, Brooks DA. Prediction of Prostate Cancer Biochemical and Clinical Recurrence Is Improved by IHC-Assisted Grading Using Appl1, Sortilin and Syndecan-1. Cancers (Basel) 2023; 15:3215. [PMID: 37370825 DOI: 10.3390/cancers15123215] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Gleason scoring is used within a five-tier risk stratification system to guide therapeutic decisions for patients with prostate cancer. This study aimed to compare the predictive performance of routine H&E or biomarker-assisted ISUP (International Society of Urological Pathology) grade grouping for assessing the risk of biochemical recurrence (BCR) and clinical recurrence (CR) in patients with prostate cancer. This retrospective study was an assessment of 114 men with prostate cancer who provided radical prostatectomy samples to the Australian Prostate Cancer Bioresource between 2006 and 2014. The prediction of CR was the primary outcome (median time to CR 79.8 months), and BCR was assessed as a secondary outcome (median time to BCR 41.7 months). The associations of (1) H&E ISUP grade groups and (2) modified ISUP grade groups informed by the Appl1, Sortilin and Syndecan-1 immunohistochemistry (IHC) labelling were modelled with BCR and CR using Cox proportional hazard approaches. IHC-assisted grading was more predictive than H&E for BCR (C-statistic 0.63 vs. 0.59) and CR (C-statistic 0.71 vs. 0.66). On adjusted analysis, IHC-assisted ISUP grading was independently associated with both outcome measures. IHC-assisted ISUP grading using the biomarker panel was an independent predictor of individual BCR and CR. Prospective studies are needed to further validate this biomarker technology and to define BCR and CR associations in real-world cohorts.
Collapse
Affiliation(s)
- Jessica M Logan
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA 5042, Australia
| | - Carmela Martini
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Alexandra Sorvina
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Prerna Tewari
- Department of Histopathology, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Sarita Prabhakaran
- Department of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Chelsea Huzzell
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Ian R D Johnson
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Ben S-Y Ung
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Joanna Lazniewska
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Robert D Brooks
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Courtney R Moore
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Maria C Caruso
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Litsa Karageorgos
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Cara M Martin
- Department of Histopathology, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | | | - Mark P Ward
- Department of Histopathology, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Mark Bates
- Department of Histopathology, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Melbourne, VIC 3001, Australia
| | - Adrian Esterman
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| | - Sheena Heffernan
- Department of Pathology, The Coombe Women and Infants University Hospital, D08 XW7X Dublin, Ireland
| | - Helen Keegan
- Department of Histopathology, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Sarah Ní Mhaolcatha
- Department of Pathology, The Coombe Women and Infants University Hospital, D08 XW7X Dublin, Ireland
| | - Roisin O'Connor
- Department of Pathology, The Coombe Women and Infants University Hospital, D08 XW7X Dublin, Ireland
| | - Victoria Malone
- Department of Pathology, The Coombe Women and Infants University Hospital, D08 XW7X Dublin, Ireland
| | - Marguerite Carter
- Department of Pathology, The Coombe Women and Infants University Hospital, D08 XW7X Dublin, Ireland
| | - Katie Ryan
- Department of Pathology, The Coombe Women and Infants University Hospital, D08 XW7X Dublin, Ireland
| | - Andres Clarke
- Department of Pathology, The Coombe Women and Infants University Hospital, D08 XW7X Dublin, Ireland
| | - Nathan Brady
- Department of Pathology, The Coombe Women and Infants University Hospital, D08 XW7X Dublin, Ireland
| | - Sonja Klebe
- Department of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Hemamali Samaratunga
- Aquesta Uropathology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Brett Delahunt
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Michael J Sorich
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA 5042, Australia
| | - Kim Moretti
- Discipline of Surgery, University of Adelaide, Adelaide, SA 5371, Australia
- Allied Health and Human Performance, University of South Australia, Adelaide, SA 5005, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Lisa M Butler
- South Australian ImmunoGENomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA 5005, Australia
- Solid Tumour Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, Bradley Building, City West Campus, North Terrace, Adelaide, SA 5000, Australia
| |
Collapse
|
32
|
Kiełb P, Kowalczyk K, Gurwin A, Nowak Ł, Krajewski W, Sosnowski R, Szydełko T, Małkiewicz B. Novel Histopathological Biomarkers in Prostate Cancer: Implications and Perspectives. Biomedicines 2023; 11:1552. [PMID: 37371647 DOI: 10.3390/biomedicines11061552] [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: 03/29/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer (PCa) is the second most frequently diagnosed cancer in men. Despite the significant progress in cancer diagnosis and treatment over the last few years, the approach to disease detection and therapy still does not include histopathological biomarkers. The dissemination of PCa is strictly related to the creation of a premetastatic niche, which can be detected by altered levels of specific biomarkers. To date, the risk factors for biochemical recurrence include lymph node status, prostate-specific antigen (PSA), PSA density (PSAD), body mass index (BMI), pathological Gleason score, seminal vesicle invasion, extraprostatic extension, and intraductal carcinoma. In the future, biomarkers might represent another prognostic factor, as discussed in many studies. In this review, we focus on histopathological biomarkers (particularly CD169 macrophages, neuropilin-1, cofilin-1, interleukin-17, signal transducer and activator of transcription protein 3 (STAT3), LIM domain kinase 1 (LIMK1), CD15, AMACR, prostate-specific membrane antigen (PSMA), Appl1, Sortilin, Syndecan-1, and p63) and their potential application in decision making regarding the prognosis and treatment of PCa patients. We refer to studies that found a correlation between the levels of biomarkers and tumor characteristics as well as clinical outcomes. We also hypothesize about the potential use of histopathological markers as a target for novel immunotherapeutic drugs or targeted radionuclide therapy, which may be used as adjuvant therapy in the future.
Collapse
Affiliation(s)
- Paweł Kiełb
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Kamil Kowalczyk
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Adam Gurwin
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Łukasz Nowak
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Wojciech Krajewski
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Roman Sosnowski
- Department of Urogenital Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Tomasz Szydełko
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| | - Bartosz Małkiewicz
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wrocław Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
33
|
Shafique A, Brughera M, Lualdi M, Alberio T. The Role of Rab Proteins in Mitophagy: Insights into Neurodegenerative Diseases. Int J Mol Sci 2023; 24:6268. [PMID: 37047239 PMCID: PMC10094445 DOI: 10.3390/ijms24076268] [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: 03/06/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Mitochondrial dysfunction and vesicular trafficking alterations have been implicated in the pathogenesis of several neurodegenerative diseases. It has become clear that pathogenetic pathways leading to neurodegeneration are often interconnected. Indeed, growing evidence suggests a concerted contribution of impaired mitophagy and vesicles formation in the dysregulation of neuronal homeostasis, contributing to neuronal cell death. Among the molecular factors involved in the trafficking of vesicles, Ras analog in brain (Rab) proteins seem to play a central role in mitochondrial quality checking and disposal through both canonical PINK1/Parkin-mediated mitophagy and novel alternative pathways. In turn, the lack of proper elimination of dysfunctional mitochondria has emerged as a possible causative/early event in some neurodegenerative diseases. Here, we provide an overview of major findings in recent years highlighting the role of Rab proteins in dysfunctional mitochondrial dynamics and mitophagy, which are characteristic of neurodegenerative diseases. A further effort should be made in the coming years to clarify the sequential order of events and the molecular factors involved in the different processes. A clear cause-effect view of the pathogenetic pathways may help in understanding the molecular basis of neurodegeneration.
Collapse
Affiliation(s)
| | | | | | - Tiziana Alberio
- Department of Science and High Technology, Center of Research in Neuroscience, University of Insubria, I-21052 Busto Arsizio, VA, Italy
| |
Collapse
|
34
|
Liao S, Lin Y, Liu L, Yang S, Lin Y, He J, Shao Y. ADAM10-a "multitasker" in sepsis: focus on its posttranslational target. Inflamm Res 2023; 72:395-423. [PMID: 36565333 PMCID: PMC9789377 DOI: 10.1007/s00011-022-01673-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 07/25/2022] [Accepted: 11/30/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Sepsis has a complex pathogenesis in which the uncontrolled systemic inflammatory response triggered by infection leads to vascular barrier disruption, microcirculation dysfunction and multiple organ dysfunction syndrome. Numerous recent studies reveal that a disintegrin and metalloproteinase 10 (ADAM10) acts as a "molecular scissor" playing a pivotal role in the inflammatory response during sepsis by regulating proteolysis by cleaving various membrane protein substrates, including proinflammatory cytokines, cadherins and Notch, which are involved in intercellular communication. ADAM10 can also act as the cellular receptor for Staphylococcus aureus α-toxin, leading to lethal sepsis. However, its substrate-specific modulation and precise targets in sepsis have not yet to be elucidated. METHODS We performed a computer-based online search using PubMed and Google Scholar for published articles concerning ADAM10 and sepsis. CONCLUSIONS In this review, we focus on the functions of ADAM10 in sepsis-related complex endothelium-immune cell interactions and microcirculation dysfunction through the diversity of its substrates and its enzymatic activity. In addition, we highlight the posttranslational mechanisms of ADAM10 at specific subcellular sites, or in multimolecular complexes, which will provide the insight to intervene in the pathophysiological process of sepsis caused by ADAM10 dysregulation.
Collapse
Affiliation(s)
- Shuanglin Liao
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - Yao Lin
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Lizhen Liu
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - Shuai Yang
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - YingYing Lin
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Junbing He
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Yiming Shao
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
- grid.410560.60000 0004 1760 3078The Key Laboratory of Sepsis Translational Medicine, Guangdong Medical University, Zhanjiang, Guangdong China
| |
Collapse
|
35
|
Xing J, Wang R, Cui F, Song L, Ma Q, Xu H. Role of the regulation of mesenchymal stem cells on macrophages in sepsis. Int J Immunopathol Pharmacol 2023; 37:3946320221150722. [PMID: 36840553 PMCID: PMC9969469 DOI: 10.1177/03946320221150722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
Sepsis is a common clinical critical disease with high mortality. The excessive release of cytokines from macrophages is the main cause of out-of-control immune response in sepsis. Mesenchymal stem cells (MSCs) are thought to be useful in adjunctive therapy of sepsis and related diseases, due to their function in immune regulation, anti-inflammatory, antibacterial, and tissue regeneration. Also there have been several successful cases in clinical treatment. Some previous studies have shown that MSCs regulate the function of macrophages through secreting cytokines and extracellular vesicles, or transferring mitochondria directly to target cells, which affects the progress of sepsis. Here, we review the regulation of MSCs on macrophages in sepsis, mainly focus on the regulation ways. We hope that will help to understand the immunological mechanism and also provide some clues for the clinical application of MSCs in the biotherapy of sepsis.
Collapse
Affiliation(s)
- Jie Xing
- Fenyang Hospital of Shanxi
Province, Fenyang, China
| | - Rui Wang
- School of Life Sciences, Northwestern Polytechnical
University, Xi’an, China
| | - Fengqi Cui
- School of Life Sciences, Northwestern Polytechnical
University, Xi’an, China
| | - Linlin Song
- Fenyang Hospital of Shanxi
Province, Fenyang, China
| | - Quanlin Ma
- Fenyang Hospital of Shanxi
Province, Fenyang, China,Quanlin Ma, Department of Cardiothoracic
Surgery, Fenyang Hospital of Shanxi Province, 186 Shengli Street, Fenyang
032200, China.
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical
University, Xi’an, China,Huiyun Xu, School of Life Sciences,
Northwestern Polytechnical University, 127 Youyi Xilu, Xi’an 710072, China.
| |
Collapse
|
36
|
Hua SS, Ding JJ, Sun TC, Guo C, Zhang Y, Yu ZH, Cao YQ, Zhong LH, Wu Y, Guo LY, Luo JH, Cui YH, Qiu S. NMDAR-dependent synaptic potentiation via APPL1 signaling is required for the accessibility of a prefrontal neuronal assembly in retrieving fear extinction. Biol Psychiatry 2023:S0006-3223(23)00087-2. [PMID: 36842495 DOI: 10.1016/j.biopsych.2023.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND The ventromedial prefrontal cortex (vmPFC) has been viewed as a locus to store and recall extinction memory. However, the synaptic and cellular mechanisms underlying this process remain elusive. METHODS We combined transgenic mice, electrophysiological recording, activity-dependent cell labeling, and chemogenetic manipulation to analyze the role of adaptor protein APPL1 in the vmPFC for fear extinction retrieval. RESULTS We found that both constitutive and conditional APPL1 knockout decreases NMDA receptor (NMDAR) function in the vmPFC and impairs fear extinction retrieval. Moreover, APPL1 undergoes nuclear translocation during extinction retrieval. Blocking APPL1 nucleocytoplasmic translocation reduces NMDAR currents and disrupts extinction retrieval. We further identified a prefrontal neuronal ensemble that is both necessary and sufficient for the storage of extinction memory. Inducible APPL1 knockout in this ensemble abolishes NMDAR-dependent synaptic potentiation and disrupts extinction retrieval, while simultaneously chemogenetic activation of this ensemble rescues the impaired behaviors. CONCLUSIONS Therefore, our results indicate that a prefrontal neuronal ensemble stores extinction memory, and APPL1 signaling supports these neurons to retrieve extinction memory via controlling NMDAR-dependent potentiation.
Collapse
Affiliation(s)
- Shu-Shan Hua
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jin-Jun Ding
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Tian-Cheng Sun
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chen Guo
- Department of Neurobiology and Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ying Zhang
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zi-Hui Yu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yi-Qing Cao
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lin-Hong Zhong
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yu Wu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lu-Ying Guo
- Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jian-Hong Luo
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology,ZhejiangUniversity ,Hangzhou ,310058 ,China
| | - Yi-Hui Cui
- Department of Neurobiology and Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Shuang Qiu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology,ZhejiangUniversity ,Hangzhou ,310058 ,China.
| |
Collapse
|
37
|
Chemical mimetics of the N-degron pathway alleviate systemic inflammation by activating mitophagy and immunometabolic remodeling. Exp Mol Med 2023; 55:333-346. [PMID: 36720915 PMCID: PMC9981610 DOI: 10.1038/s12276-023-00929-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/19/2022] [Accepted: 11/04/2022] [Indexed: 02/02/2023] Open
Abstract
The Arg/N-degron pathway, which is involved in the degradation of proteins bearing an N-terminal signal peptide, is connected to p62/SQSTM1-mediated autophagy. However, the impact of the molecular link between the N-degron and autophagy pathways is largely unknown in the context of systemic inflammation. Here, we show that chemical mimetics of the N-degron Nt-Arg pathway (p62 ligands) decreased mortality in sepsis and inhibited pathological inflammation by activating mitophagy and immunometabolic remodeling. The p62 ligands alleviated systemic inflammation in a mouse model of lipopolysaccharide (LPS)-induced septic shock and in the cecal ligation and puncture model of sepsis. In macrophages, the p62 ligand attenuated the production of proinflammatory cytokines and chemokines in response to various innate immune stimuli. Mechanistically, the p62 ligand augmented LPS-induced mitophagy and inhibited the production of mitochondrial reactive oxygen species in macrophages. The p62 ligand-mediated anti-inflammatory, antioxidative, and mitophagy-activating effects depended on p62. In parallel, the p62 ligand significantly downregulated the LPS-induced upregulation of aerobic glycolysis and lactate production. Together, our findings demonstrate that p62 ligands play a critical role in the regulation of inflammatory responses by orchestrating mitophagy and immunometabolic remodeling.
Collapse
|
38
|
Martini C, Logan JM, Sorvina A, Gordon C, Beck AR, S-Y Ung B, Caruso MC, Moore C, Hocking A, Johnson IRD, Li KL, Karageorgos L, Hopkins AM, Esterman AJ, Huzzell C, Brooks RD, Lazniewska J, Hickey SM, Bader C, Parkinson-Lawrence E, Weigert R, Sorich MJ, Tewari P, Martin C, O'Toole S, Bates M, Ward M, Mohammed B, Keegan H, Watson W, Prendergast S, Heffernan S, NiMhaolcatha S, O'Connor R, Malone V, Carter M, Ryan K, Brady N, Clarke A, Sokol F, Prabhakaran S, Stahl J, Klebe S, Samaratunga H, Delahunt B, Selemidis S, Moretti KL, Butler LM, O'Leary JJ, Brooks DA. Aberrant protein expression of Appl1, Sortilin and Syndecan-1 during the biological progression of prostate cancer. Pathology 2023; 55:40-51. [PMID: 36089417 DOI: 10.1016/j.pathol.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 01/27/2023]
Abstract
Diagnosis and assessment of patients with prostate cancer is dependent on accurate interpretation and grading of histopathology. However, morphology does not necessarily reflect the complex biological changes occurring in prostate cancer disease progression, and current biomarkers have demonstrated limited clinical utility in patient assessment. This study aimed to develop biomarkers that accurately define prostate cancer biology by distinguishing specific pathological features that enable reliable interpretation of pathology for accurate Gleason grading of patients. Online gene expression databases were interrogated and a pathogenic pathway for prostate cancer was identified. The protein expression of key genes in the pathway, including adaptor protein containing a pleckstrin homology (PH) domain, phosphotyrosine-binding (PTB) domain, and leucine zipper motif 1 (Appl1), Sortilin and Syndecan-1, was examined by immunohistochemistry (IHC) in a pilot study of 29 patients with prostate cancer, using monoclonal antibodies designed against unique epitopes. Appl1, Sortilin, and Syndecan-1 expression was first assessed in a tissue microarray cohort of 112 patient samples, demonstrating that the monoclonal antibodies clearly illustrate gland morphologies. To determine the impact of a novel IHC-assisted interpretation (the utility of Appl1, Sortilin, and Syndecan-1 labelling as a panel) of Gleason grading, versus standard haematoxylin and eosin (H&E) Gleason grade assignment, a radical prostatectomy sample cohort comprising 114 patients was assessed. In comparison to H&E, the utility of the biomarker panel reduced subjectivity in interpretation of prostate cancer tissue morphology and improved the reliability of pathology assessment, resulting in Gleason grade redistribution for 41% of patient samples. Importantly, for equivocal IHC-assisted labelling and H&E staining results, the cancer morphology interpretation could be more accurately applied upon re-review of the H&E tissue sections. This study addresses a key issue in the field of prostate cancer pathology by presenting a novel combination of three biomarkers and has the potential to transform clinical pathology practice by standardising the interpretation of the tissue morphology.
Collapse
Affiliation(s)
- Carmela Martini
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia.
| | - Jessica M Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Alexandra Sorvina
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Colin Gordon
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Andrew R Beck
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Ben S-Y Ung
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Maria C Caruso
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Courtney Moore
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Ashleigh Hocking
- Department of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ian R D Johnson
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Ka Lok Li
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Litsa Karageorgos
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA, Australia
| | - Adrian J Esterman
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Chelsea Huzzell
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Robert D Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Joanna Lazniewska
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Christie Bader
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | | | - Roberto Weigert
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Michael J Sorich
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA, Australia
| | - Prerna Tewari
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Cara Martin
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Mark Ward
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Bashir Mohammed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Helen Keegan
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - William Watson
- University College Dublin, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Sophie Prendergast
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Sheena Heffernan
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Sarah NiMhaolcatha
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Roisin O'Connor
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Victoria Malone
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Marguerite Carter
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Katie Ryan
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Nathan Brady
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Andres Clarke
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Filip Sokol
- Department of Pathology, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Sarita Prabhakaran
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia; Department of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Jürgen Stahl
- Department of Cytopathology and Histopathology, Clinpath Pathology, Adelaide, SA, Australia
| | - Sonja Klebe
- Department of Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Department of Surgical Pathology, SA Pathology at Flinders Medical Centre, Adelaide, SA, Australia
| | | | - Brett Delahunt
- Department of Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Melbourne, Vic, Australia
| | - Kim L Moretti
- Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia; University of South Australia, Adelaide, SA, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Vic, Australia
| | - Lisa M Butler
- South Australian ImmunoGENomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia; Solid Tumour Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
39
|
Yu T, Lu X, Liang Y, Yang L, Yin Y, Chen H. Ononin alleviates DSS-induced colitis through inhibiting NLRP3 inflammasome via triggering mitophagy. Immun Inflamm Dis 2023; 11:e776. [PMID: 36840499 PMCID: PMC9910166 DOI: 10.1002/iid3.776] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/09/2023] [Accepted: 01/13/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Ononin, a flavonoid isolated from Astragalus membranaceus root, is the active ingredient of A. membranaceus and has potential anti-inflammatory properties, but its effect on colitis is unclear. AIMS This study aimed to explore the anticolitis effect of Ononin by establishing a colitis model in mice induced by dextran sulfate sodium (DSS). METHODS Male C57BL/6 mice were provided DSS, then treated with Ononin (10, 20, 40 mg/kg) or 5-ASA (40 mg/kg). The colitis symptoms were observed, the disease activity index (DAI) score were recorded daily, and colonic inflammation was evaluted by histopathological scoring. The expression of cytokines, inflammatory mediators, and mitophagy/NLRP3 inflammasome-related proteins were measured. RESULTS Ononin significantly alleviated weight loss and colon shortening in mice with colitis (p < .01). Moreover, Ononin decreased the production of inflammatory cytokines and mediators associated with colitis (p < .05). In addition, Ononin inhibited macrophages infiltration and reduced caspase-1 activation in colitis mice. Caspase-1 activation is closely related to the NLRP3 inflammasome. Therefore, we investigated the effect of Ononin on NLRP3 inflammasome in vitro. The relevant results confirmed that Ononin inhibited NLRP3 inflammasome activation and inhibited mitochondrial damage (p < .05). Further studies revealed that Ononin inhibited mitochondrial damage through triggering mitophagy (p < .05). CONCLUSION Ononin alleviates DSS-induced colitis by activating mitophagy to inhibit NLRP3 inflammasome.
Collapse
Affiliation(s)
- Ting Yu
- Department of Gastroenterology, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingJiangsuPeople's Republic of China
| | - Xuejia Lu
- Department of Gastroenterology, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingJiangsuPeople's Republic of China
| | - Yan Liang
- Department of Gastroenterology, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingJiangsuPeople's Republic of China
| | - Lin Yang
- Department of Gastroenterology, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingJiangsuPeople's Republic of China
| | - Yuehan Yin
- China HuaYou Group CorporationBeijingPeople's Republic of China
| | - Hong Chen
- Department of Gastroenterology, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingJiangsuPeople's Republic of China
| |
Collapse
|
40
|
Li C, Ma J, Mali N, Zhang L, Wei T, Shi L, Liu F, WenXing F, Yang J. Relevance of the pyroptosis-related inflammasome drug targets in the Chuanxiong to improve diabetic nephropathy. Mol Med 2022; 28:136. [PMID: 36401196 PMCID: PMC9673343 DOI: 10.1186/s10020-022-00567-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022] Open
Abstract
Background A chronic inflammatory disease caused by disturbances in metabolism, diabetic nephropathy (DN) is a chronic inflammatory disease. Pyroptosis is a novel form of programmed cell death in many inflammation-related diseases, including DN. Therefore, pyroptosis could be a promising target for DN therapy. Methods To get the components and pharmacodynamic targets of Chuanxiong, we identified by searching TCMID, TCMSP, ETCM and HERB databases. Then, from the Molecular Signatures Database (MSigDB) and Gene Ontology (GO) database, pyroptosis genes were collected. Identification of critical genes in DN by bioinformatics analysis and then using the ConsensusClusterPlus package to divide the express data of diff genes into some subgroups with different levels of pyroptosis; the WGCNA machine algorithm was used to simulate the mechanism Chuanxiong improving DN. Results In this study, we found DHCR24, ANXA1, HMOX1, CDH13, ALDH1A1, LTF, CHI3L1, CACNB2, and MTHFD2 interacted with the diff genes of DN. We used GSE96804 as a validation set to evaluate the changes of APIP, CASP6, CHMP2B, CYCS, DPP8, and TP53 in four different cell proapoptotic states. WGCNA analysis showed that DHCR24, CHI3L1, and CACNB2 had significant changes in different cell proapoptotic levels. In the experimental stage, we also confirmed that the active ingredients of Chuanxiong could improve the inflammatory state and the levels of pyroptosis under high glucose. Conclusion The improvement of DN by Chuanxiong is related to the change of pyroptosis.
Collapse
|
41
|
Xu F, Wu Y, Yang Q, Cheng Y, Xu J, Zhang Y, Dai H, Wang B, Ma Q, Chen Y, Lin F, Wang C. Engineered Extracellular Vesicles with SHP2 High Expression Promote Mitophagy for Alzheimer's Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2207107. [PMID: 36193769 DOI: 10.1002/adma.202207107] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/18/2022] [Indexed: 06/16/2023]
Abstract
Mitochondrial dysfunction is a fundamental pathological feature of Alzheimer's disease (AD). However, toxicity and poor brain enrichment of existing mitophagy inducers limit their further applications. In this study, a platform for AD therapy is developed using nanosized mesenchymal-stem-cells-derived extracellular vesicles with tyrosine phosphatase-2 (SHP2) high-expression (MSC-EVs-SHP2). The high blood-brain barrier penetration ability of MSC-EVs-SHP2 is demonstrated in AD-mice, facilitating SHP2 delivery to the brain. In addition, MSC-EVs-SHP2 significantly induces mitophagy of neuronal cells, which alleviates mitochondrial damage-mediated apoptosis and NLRP3 inflammasome activation. Mitophagy further diminishes neuronal cells apoptosis and neuroinflammation, culminating with rescued synaptic loss and cognitive decline in an AD mouse model. The EV-engineering technology provides a potential platform for effective AD therapy by inducing mitophagy.
Collapse
Affiliation(s)
- Fang Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yi Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Qianyu Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Ying Cheng
- Institute of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Disease, College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, 215123, P. R. China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yue Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Beilei Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Qingle Ma
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yitong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Fang Lin
- Institute of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Disease, College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, 215123, P. R. China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| |
Collapse
|
42
|
SS-31 Improves Cognitive Function in Sepsis-Associated Encephalopathy by Inhibiting the Drp1-NLRP3 Inflammasome Activation. Neuromolecular Med 2022:10.1007/s12017-022-08730-1. [DOI: 10.1007/s12017-022-08730-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
43
|
Jiang H, Chen F, Song D, Zhou X, Ren L, Zeng M. Dynamin-Related Protein 1 Is Involved in Mitochondrial Damage, Defective Mitophagy, and NLRP3 Inflammasome Activation Induced by MSU Crystals. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5064494. [PMID: 36338340 PMCID: PMC9627272 DOI: 10.1155/2022/5064494] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/21/2022] [Indexed: 12/02/2023]
Abstract
Excessive generation of reactive oxygen species (ROS) has great impacts on MSU crystal-induced inflammation. Drp1-dependent mitochondrial fission is closely associated with mitochondrial ROS levels. However, whether Drp1 signaling contributes to MSU crystal-induced inflammation remains unclear. Mice bone marrow-derived macrophages (BMDMs) were primed with LPS and then stimulated with MSU suspensions for 12 h. The protein levels associated with mitochondrial dynamics, oxidative stress, and mitophagy were detected by Western blot. BMDMs were loaded with MitoTracker Green probe to detect mitochondrial morphology. To measure mitochondrial reactive oxygen species (ROS) and total ROS levels, cells were loaded, respectively, with MitoSOX and DHE probes. The effects of Mito-TEMPO, an antioxidant that targets the mitochondria or DRP1 inhibitor (Mdivi-1) on MSU crystal-induced peritonitis and arthritis mouse models, were evaluated. Our study revealed that MSU crystal stimulation resulted in elevation of mitochondrial fragmentation of BMDMs. Treatment with Mito-TEMPO or Drp1 knockdown significantly ameliorated the mitochondrial damage induced by MSU crystals. BMDMs exposure to MSU crystals increased the expression of auto/mitophagy marker proteins and promoted the fusion of mitophagosomes with lysosomes, leading to accumulation of mitolysosomes. Drp1 knockdown alleviated defective mitophagy and activation of the NLRP3 inflammasome in MSU crystal-treated BMDMs. This study indicates that there is crosstalk between mitochondrial ROS and Drp1 signaling in MSU crystal-induced inflammation. Drp1 signaling is involved in MSU crystal-induced mitochondrial damage, impaired mitophagy and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Hui Jiang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
- Medical Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
| | - Feng Chen
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
- Medical Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
| | - DianZe Song
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
- Medical Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
| | - Xiaoqin Zhou
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
| | - Long Ren
- The Fifth People's Hospital of Nanchong City, 21# Bajiao Street, Nanchong, 637100 Sichuan, China
| | - Mei Zeng
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College and Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
- Medical Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Nanchong, 637001 Sichuan, China
- The Fifth People's Hospital of Nanchong City, 21# Bajiao Street, Nanchong, 637100 Sichuan, China
| |
Collapse
|
44
|
Hu H, Guo L, Overholser J, Wang X. Mitochondrial VDAC1: A Potential Therapeutic Target of Inflammation-Related Diseases and Clinical Opportunities. Cells 2022; 11:cells11193174. [PMID: 36231136 PMCID: PMC9562648 DOI: 10.3390/cells11193174] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 12/03/2022] Open
Abstract
The multifunctional protein, voltage-dependent anion channel 1 (VDAC1), is located on the mitochondrial outer membrane. It is a pivotal protein that maintains mitochondrial function to power cellular bioactivities via energy generation. VDAC1 is involved in regulating energy production, mitochondrial oxidase stress, Ca2+ transportation, substance metabolism, apoptosis, mitochondrial autophagy (mitophagy), and many other functions. VDAC1 malfunction is associated with mitochondrial disorders that affect inflammatory responses, resulting in an up-regulation of the body’s defensive response to stress stimulation. Overresponses to inflammation may cause chronic diseases. Mitochondrial DNA (mtDNA) acts as a danger signal that can further trigger native immune system activities after its secretion. VDAC1 mediates the release of mtDNA into the cytoplasm to enhance cytokine levels by activating immune responses. VDAC1 regulates mitochondrial Ca2+ transportation, lipid metabolism and mitophagy, which are involved in inflammation-related disease pathogenesis. Many scientists have suggested approaches to deal with inflammation overresponse issues via specific targeting therapies. Due to the broad functionality of VDAC1, it may become a useful target for therapy in inflammation-related diseases. The mechanisms of VDAC1 and its role in inflammation require further exploration. We comprehensively and systematically summarized the role of VDAC1 in the inflammatory response, and hope that our research will lead to novel therapeutic strategies that target VDAC1 in order to treat inflammation-related disorders.
Collapse
Affiliation(s)
- Hang Hu
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Linlin Guo
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center at The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (L.G.); (X.W.)
| | - Jay Overholser
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center at The Ohio State University, Columbus, OH 43210, USA
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Correspondence: (L.G.); (X.W.)
| |
Collapse
|
45
|
Irisin Promotes Osteogenesis by Modulating Oxidative Stress and Mitophagy through SIRT3 Signaling under Diabetic Conditions. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3319056. [PMID: 36262283 PMCID: PMC9576424 DOI: 10.1155/2022/3319056] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
Advanced glycation end products (AGEs) accumulate in the bone tissue of patients with diabetes mellitus, resulting in oxidative stress, poor bone healing, or regeneration. Irisin, a novel exercise-induced myokine, is involved in the regulation of bone metabolism. However, the effects of irisin on adipose-derived stem cell (ASC) osteogenic differentiation and bone healing under diabetic conditions remain poorly understood. ASCs were obtained from inguinal fat of Sprague-Dawley rats and treated with different concentrations of AGEs and irisin. Cell proliferation, apoptosis, and osteogenic differentiation abilities of ASCs were detected. To explore the regulatory role of sirtuin 3 (SIRT3), ASCs were transfected with lentivirus-mediated SIRT3 overexpression or knockdown vectors. Next, we investigated mitochondrial functions, mitophagy, and mitochondrial biogenesis in different groups. Moreover, SOD2 acetylation and potential signaling pathways were assessed. Additionally, a diabetic rat model was used to evaluate the effect of irisin on bone healing in calvarial critical-sized defects (CSDs) in vivo. Our results showed that irisin incubation mitigated the inhibitory effects of AGEs on ASCs by increasing cell viability and promoting osteogenesis. Moreover, irisin modulated mitochondrial membrane potential, intracellular ROS levels, mitochondrial O2·− status, ATP generation, complex I and IV activities, mitophagy, and mitochondrial biogenesis via a SIRT3-mediated pathway under AGEs exposure. Furthermore, in calvarial CSDs of diabetic rats, transplantation of gels encapsulating irisin-pretreated ASCs along with irisin largely enhanced bone healing. These findings suggest that irisin attenuates AGE-induced ASC dysfunction through SIRT3-mediated maintenance of oxidative stress homeostasis and regulation of mitophagy and mitochondrial biogenesis. Thus, our studies shed new light on the role of irisin in promoting the ASC osteogenesis and targeting SIRT3 as a novel therapeutic intervention strategy for bone regeneration under diabetic conditions.
Collapse
|
46
|
Zhang Y, Liu W, Yuan W, Cai Z, Ye G, Zheng G, Xu C, Wang X, Zeng C, Mi R, Feng P, Chen F, Wu Y, Shen H, Wang P. Impairment of APPL1/Myoferlin facilitates adipogenic differentiation of mesenchymal stem cells by blocking autophagy flux in osteoporosis. Cell Mol Life Sci 2022; 79:488. [PMID: 35984564 PMCID: PMC9391247 DOI: 10.1007/s00018-022-04511-y] [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: 05/24/2022] [Revised: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022]
Abstract
An imbalance of human mesenchymal stem cells (hMSCs) adipogenic and osteogenic differentiation is crucial in the pathogenesis of osteoporosis, and elucidation of the underlying mechanism is urgently needed. APPL1, an adaptor protein of the adiponectin receptor, was recently shown to be closely related to bone mass. However, the role of APPL1 in the imbalance of hMSC differentiation in osteoporosis is unclear. Therefore, we aimed to explore the mechanisms by which APPL1 alters hMSCs adipogenic differentiation in osteoporosis. Here, we found that APPL1 expression was downregulated in elderly patients with osteoporosis and in mouse osteoporosis model. APPL1 negatively regulated hMSC adipogenic differentiation in vivo and in vitro. Mechanistically, by enhancing ubiquitination-mediated Myoferlin degradation, downregulated APPL1 expression increased the risk of lysosome dysfunction during hMSCs adipogenic differentiation. Lysosomal dysfunction inhibited autophagy flux by suppressing autophagosome degradation and promoted hMSC differentiation towards the adipocyte lineage. Our findings suggest that APPL1/Myoferlin downregulation promoted hMSCs adipogenic differentiation by inhibiting autophagy flux, further impairing the balance of hMSCs adipogenic and osteogenic differentiation in osteoporosis; the APPL1/ Myoferlin axis may be a promising diagnostic and therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Yunhui Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Weiquan Yuan
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Zhaopeng Cai
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Guiwen Ye
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Chenhao Xu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Xinglang Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Chenying Zeng
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Rujia Mi
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Pei Feng
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Fenglei Chen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China.
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025# Shennan Road, Shenzhen, 518000, People's Republic of China.
| |
Collapse
|
47
|
Hormesis and Oxidative Distress: Pathophysiology of Reactive Oxygen Species and the Open Question of Antioxidant Modulation and Supplementation. Antioxidants (Basel) 2022; 11:antiox11081613. [PMID: 36009331 PMCID: PMC9405171 DOI: 10.3390/antiox11081613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Alterations of redox homeostasis leads to a condition of resilience known as hormesis that is due to the activation of redox-sensitive pathways stimulating cell proliferation, growth, differentiation, and angiogenesis. Instead, supraphysiological production of reactive oxygen species (ROS) exceeds antioxidant defence and leads to oxidative distress. This condition induces damage to biomolecules and is responsible or co-responsible for the onset of several chronic pathologies. Thus, a dietary antioxidant supplementation has been proposed in order to prevent aging, cardiovascular and degenerative diseases as well as carcinogenesis. However, this approach has failed to demonstrate efficacy, often leading to harmful side effects, in particular in patients affected by cancer. In this latter case, an approach based on endogenous antioxidant depletion, leading to ROS overproduction, has shown an interesting potential for enhancing susceptibility of patients to anticancer therapies. Therefore, a deep investigation of molecular pathways involved in redox balance is crucial in order to identify new molecular targets useful for the development of more effective therapeutic approaches. The review herein provides an overview of the pathophysiological role of ROS and focuses the attention on positive and negative aspects of antioxidant modulation with the intent to find new insights for a successful clinical application.
Collapse
|
48
|
Qiu Y, Huang Y, Chen M, Yang Y, Li X, Zhang W. Mitochondrial DNA in NLRP3 inflammasome activation. Int Immunopharmacol 2022; 108:108719. [DOI: 10.1016/j.intimp.2022.108719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/26/2022] [Accepted: 03/17/2022] [Indexed: 12/20/2022]
|
49
|
Wu KKL, Cheng KKY. A new role of the early endosome in restricting NLRP3 inflammasome via mitophagy. Autophagy 2022; 18:1475-1477. [PMID: 35196181 PMCID: PMC9225602 DOI: 10.1080/15548627.2022.2040314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
NLRP3 (NLR family pyrin domain containing 3) inflammasome is a potent mediator of inflammation due to its ability to produce the pro-inflammatory cytokines IL1B (interleukin 1 beta) and IL18 in response to numerous danger signals and pathogens. Mitophagy, a selective form of autophagy, restricts NLRP3 inflammasome activation by limiting the mitochondrial-derived danger signals. Here, we demonstrated that the adaptor protein APPL1 together with its interaction partner RAB5 in early endosomes negatively regulate NLRP3 inflammasome activation via induction of mitophagy in macrophages. Hematopoietic-deletion of Appl1 exacerbates systemic NLRP3 inflammasome activation in rodent models under obese or septic conditions. Our study identified a new regulatory network between early endosomes and mitochondria in control of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Kelvin Ka Lok Wu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Kenneth King Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China,CONTACT Kenneth King Yip Cheng Department of Health Technology and Informatics, he Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| |
Collapse
|