1
|
Liu S, Xu Y, Yao X, Cao H, Zhou H, Luo J, Gao H, Chen B, Chen H, Xie T, Zhan X. Perillaldehyde ameliorates sepsis-associated acute kidney injury via inhibiting HSP90AA1-mediated ferroptosis and pyroptosis: Molecular structure and protein interaction of HSP90AA1. Int J Biol Macromol 2025; 304:140954. [PMID: 39947536 DOI: 10.1016/j.ijbiomac.2025.140954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/16/2025]
Abstract
Heat shock protein 90α (HSP90AA1) is a molecular chaperone involved in a variety of cellular processes. Special attention is paid to how perillaldehyde ameliorates kidney injury by inhibiting HSP90AA1-mediated iron and pyrotoxicity, and in-depth analysis of the molecular structure and protein interactions of HSP90AA-1. The interaction between perillaldehyde and HSP90AA1 and the effect of perillaldehyde on the molecular structure of HSP90AA1 were analyzed by molecular docking and surface plasmon resonance technique. Western blot and immunohistochemical results showed that perillaldehyde could decrease the expression of HSP90AA1 and change its distribution in the kidney. Molecular docking and surface plasmonic resonance experiments revealed the high affinity binding between perillaldehyde and HSP90AA1, and further analysis showed that perillaldehyde could induce the conformational change of HSP90AA1, thereby inhibiting its function.
Collapse
Affiliation(s)
- Shuai Liu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yunfei Xu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Heng Cao
- Department of Urology, The Third the People's Hospital of Bengbu, Bengbu Medical College, Bengbu 233000, China
| | - Hongmin Zhou
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jun Luo
- Department of Urology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Hanlu Gao
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Bowen Chen
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Hao Chen
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Tiancheng Xie
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xiangcheng Zhan
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
2
|
Cui Z, He T, Zhang S. The efficient prediction of inflammatory osteolysis caused by polylactic acid through network toxicology and molecular docking strategy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117876. [PMID: 39947065 DOI: 10.1016/j.ecoenv.2025.117876] [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: 10/23/2024] [Revised: 01/16/2025] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Polylactic acid (PLA), as a bioplastic, is extensively utilized in bone tissue engineering for its biocompatibility, adaptability and affordability. However, the toxicological research of PLA is still limited. The hydrolysis products of PLA induced inflammatory response which caused inflammatory osteolysis mediated by oxidative damage through the recruitment of macrophages and the accumulation of foreign body multinucleated giant cells, ultimately leading to the failure of bone tissue regeneration. The lack of effective treatments highlights the importance of finding new therapies. This study systematically investigated the potential molecular mechanisms of PLA-induced inflammatory osteolysis by employing network toxicology and molecular docking techniques. We first conducted a network toxicology-based assessment according to the molecular structure of PLA. The result from integrating and screening targets from multiple databases identified 126 potential targets associated with PLA-induced inflammatory osteolysis, and then an interaction network diagram of the targets was constructed. Gene ontology (GO)/KEGG enrichment analysis clarified that PLA may cause inflammatory osteolysis via metabolic pathways and pathways in cancer, as well as lipid and atherosclerosis. Further analysis by STRING and Cytoscape software screened 25 core targets including HSP90AA1, AKT1, SRC, STAT1 and FYN. We found that the enriched highly correlated pathways covered 18 of the 25 core targets, supporting the scientific hypothesis that PLA induces inflammatory osteolysis. Moreover, the results of molecular docking confirmed that PLA displayed a strong binding ability with the core targets and formed stable binding. Taken together, this study not only revealed the potential biological mechanism of PLA-induced inflammatory osteolysis, but also provided new evidence for the future prevention and treatment of PLA-induced inflammation.
Collapse
Affiliation(s)
- Zichen Cui
- Department of thoracic surgery, The Affiliated Hospital of Qingdao University, Qingdao 266700, PR China.
| | - Tian He
- Department of Orthopedics Surgery, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264400, PR China.
| | - Shuo Zhang
- Department of thoracic surgery, The Affiliated Hospital of Qingdao University, Qingdao 266700, PR China.
| |
Collapse
|
3
|
Yang J, He B, Dang L, Liu J, Liu G, Zhao Y, Yu P, Wang Q, Wang L, Xin W. Celastrol Regulates the Hsp90-NLRP3 Interaction to Alleviate Rheumatoid Arthritis. Inflammation 2025; 48:346-360. [PMID: 38874810 DOI: 10.1007/s10753-024-02060-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: 04/03/2024] [Revised: 05/08/2024] [Accepted: 05/21/2024] [Indexed: 06/15/2024]
Abstract
Previous studies have verified that celastrol (Cel) protects against rheumatoid arthritis (RA) by inhibiting the NLRP3 inflammasome signaling pathway, but the molecular mechanism by which Cel regulates NLRP3 has not been clarified. This study explored the specific mechanisms of Cel in vitro and in vivo. A type II collagen-induced arthritis (CIA) mouse model was used to study the antiarthritic activity of Cel; analysis of paw swelling, determination of the arthritis score, and pathological examinations were performed. The antiproliferative and antimigratory effects of Cel on TNF-α induced fibroblast-like synoviocytes (FLSs) were tested. Proinflammatory factors were evaluated using enzyme-linked immunosorbent assay (ELISA). The expression of NF-κB/NLRP3 pathway components was determined by western blotting and immunofluorescence staining in vitro and in vivo. The putative binding sites between Cel and Hsp90 were predicted through molecular docking, and the binding interactions were determined using the Octet RED96 system and coimmunoprecipitation. Cel decreased arthritis severity and reduced TNF-α-induced FLSs migration and proliferation. Additionally, Cel inhibited NF-κB/NLRP3 signaling pathway activation, reactive oxygen species (ROS) production, and proinflammatory cytokine secretion. Furthermore, Cel interacted directly with Hsp90 and blocked the interaction between Hsp90 and NLRP3 in FLSs. Our findings revealed that Cel regulates NLRP3 inflammasome signaling pathways both in vivo and in vitro. These effects are induced through FLSs inhibition of the proliferation and migration by blocking the interaction between Hsp90 and NLRP3.
Collapse
Affiliation(s)
- Junjie Yang
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Biyao He
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Longjiao Dang
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Jiayu Liu
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Guohao Liu
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Yuwei Zhao
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Pengfei Yu
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Qiaoyun Wang
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Lei Wang
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Wenyu Xin
- Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
4
|
Cai H, Li H, Xiao X, Wang S, Liu R, Qin Y, Zhou Y, Yao C. TRAF6 promotes abdominal aortic aneurysm development by activating macrophage pyroptosis via the NLRP3/Caspase1/GSDMD pathway. FASEB J 2025; 39:e70318. [PMID: 39831511 DOI: 10.1096/fj.202402873r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Abdominal aortic aneurysm represents a critical pathology of the aorta that currently lacks effective pharmacological interventions. TNF receptor-associated factor 6 (TRAF6) has been established to be involved in cardiovascular diseases such as atherosclerosis, hypertension, and heart failure. However, its role in abdominal aortic aneurysm (AAA) remains unclear. This study aimed to explore the role of TRAF6 on AAA formation and its underlying mechanisms. Single-cell RNA sequencing of human AAA tissues demonstrated that TRAF6 was significantly upregulated in aortic macrophages. Moreover, overexpression of TRAF6 promotes AAA formation in elastase-induced C57BL/6 mice, while TRAF6 pharmacological inhibition could attenuate AAA development. Consistently, inhibition of TRAF6 in macrophages through in vitro methods notably limits their pyroptosis, while also diminishing proinflammatory responses in these cells. Mechanistically, TRAF6 can modulate macrophage pyroptosis through the NLRP3/Caspase1/GSDMD signaling pathway. Our study highlights the crucial role of the TRAF6/NLRP3/Caspase1/GSDMD axis in macrophage pyroptosis and AAA, offering potential biomarkers and therapeutic targets for AAA.
Collapse
Affiliation(s)
- Huoying Cai
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Huaming Li
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaoyong Xiao
- Department of Emergency Medicine, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Siwen Wang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Ruiming Liu
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Yuansen Qin
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Yu Zhou
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| | - Chen Yao
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Isshiki T, Sunakawa M, Vierhout M, Ayoub A, Ali P, Naiel S, Miyoshi S, Naqvi A, Hambly N, Kishi K, Ask K, Kolb MRJ. Upregulation of HSP90α in the lungs and circulation in sarcoidosis. Front Med (Lausanne) 2025; 12:1532437. [PMID: 39881845 PMCID: PMC11774731 DOI: 10.3389/fmed.2025.1532437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/03/2025] [Indexed: 01/31/2025] Open
Abstract
Background Sarcoidosis is a systemic granulomatous disease of unknown cause. Natural improvement with favorable outcome is common, but a significant number of patients present with difficult to manage and progressive disease. The identification of biomarkers associated with disease activity and progression is warranted. Extracellular heat shock protein 90 (HSP90) α is a signaling molecule released by cells that induces proinflammatory signaling through interaction with certain receptors, such as lipoprotein receptor-related protein 1. Materials and methods HSP90α protein expression in lung tissues derived from patients diagnosed with sarcoidosis and control subjects was assessed by immunohistochemistry. Serum HSP90α concentration was measured in sarcoidosis patients and healthy controls and correlated with clinical outcomes. Bronchoalveolar lavage fluid (BALF) was collected and analyzed for HSP90α expression. Extracellular HSP90α released from macrophages was examined in human primary cells and an immortalized cell line. Results Macrophages and granulomas in sarcoidosis-affected lungs showed high HSP90α expression. Serum HSP90α levels were elevated in sarcoidosis patients compared with controls and correlated with BALF HSP90α levels. HSP90α concentrations in the circulation were correlated with biomarkers of disease stage. Both primary and immortalized macrophages showed a high capacity for secreting extracellular HSP90α. Conclusion These results demonstrate that macrophages in the lungs of sarcoidosis patients produce high levels of HSP90α, suggesting HSP90α as a potential biomarker and therapeutic target.
Collapse
Affiliation(s)
- Takuma Isshiki
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada
- Department of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Motoko Sunakawa
- Department of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Megan Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada
| | - Anmar Ayoub
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada
| | - Pareesa Ali
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada
| | - Safaa Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada
| | - Shion Miyoshi
- Department of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Asghar Naqvi
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Nathan Hambly
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Kazuma Kishi
- Department of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, Hamilton, ON, Canada
| | - Martin R. J. Kolb
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
6
|
Jiang L, Tian CB, Ye RH, Shi N, He XC, Zhao YL, Luo XD. Kakuol and asarinin protecting liver injury via HSP90AA1/CDK2/mTOR signaling pathway. Fitoterapia 2025; 180:106297. [PMID: 39551106 DOI: 10.1016/j.fitote.2024.106297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/30/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024]
Abstract
Drug-induced liver injury caused acute hepatic failure and hepatitis frequently. In this investigation, kakuol and asarinin reduced the levels of serum alanine transaminase (ALT), aspartate transaminase (AST) and malondialdehyde (MDA) dramatically, and ameliorated the pathological damage of liver tissues in APAP-induced mice. Furthermore, both compounds increased the viabilities of APAP-induced L-O2 cells and extracellular glutathione (GSH) levels accompanied significantly by reducing the level of intracellular ROS in vitro. In addition, HSP90AA1/CDK2/mTOR signaling pathway and five target proteins (CDK2, HSP90AA1, HRAS, MMP1, mTOR) were proposed from network pharmacology and molecular docking prediction, and then the up-regulation of protein expression of CDK2, mTOR and down-regulation of HSP90AA1, HRAS, MMP1 by kakuol and asarinin in western blotting supported their mechanism.
Collapse
Affiliation(s)
- Ling Jiang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Cai-Bo Tian
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Rui-Han Ye
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Nian Shi
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Xing-Chao He
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China.
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, PR China.
| |
Collapse
|
7
|
Zhao YX, Sun YY, Li LY, Li XF, Li HD, Chen X, Xia R, Yang YL, Jiang XY, Zuo LQ, Meng XM, Wang H, Huang C, Li J. Rab11b promotes M1-like macrophage polarization by restraining autophagic degradation of NLRP3 in alcohol-associated liver disease. Acta Pharmacol Sin 2025; 46:134-146. [PMID: 38992121 PMCID: PMC11695811 DOI: 10.1038/s41401-024-01333-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/02/2024] [Indexed: 07/13/2024]
Abstract
Macrophage polarization is vital to mounting a host defense or repairing tissue in various liver diseases. Excessive activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome is related to the orchestration of inflammation and alcohol-associated liver disease (ALD) pathology. Rab GTPases play critical roles in regulating vesicular transport. In this study we investigated the role of Rab11b in ALD, aiming to identify effective therapeutic targets. Here, we first demonstrated a decreased expression of Rab11b in macrophages from ALD mice. Knockdown of Rab11b by macrophage-specific adeno-associated virus can alleviate alcohol induced liver inflammation, injury and steatosis. We found that LPS and alcohol stimulation promoted Rab11b transferring from the nucleus to the cytoplasm in bone marrow-derived macrophages (BMDM) cells. Rab11b specifically activated the NLRP3 inflammasome in BMDMs and RAW264.7 cells to induce M1 macrophage polarization. Rab11b overexpression in BMDMs inhibited autophagic flux, leading to the suppression of LC3B-mediated NLRP3 degradation. We conclude that impaired Rab11b could alleviate alcohol-induced liver injury via autophagy-mediated NLRP3 degradation.
Collapse
Affiliation(s)
- Yu-Xin Zhao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei, 230032, China
| | - Ying-Yin Sun
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Liang-Yun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei, 230032, China
| | - Xiao-Feng Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Hai-di Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei, 230032, China
| | - Xin Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei, 230032, China
| | - Ran Xia
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Ying-Li Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei, 230032, China
| | - Xin-Yu Jiang
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, 230032, China
| | - Long-Quan Zuo
- Department of Pharmacy, Hospital of Armed Police of Anhui Province, Hefei, 230032, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
- Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei, 230032, China.
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
- Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
8
|
Zhao Y, Park JY, Yang D, Zhang M. A computational framework to in silico screen for drug-induced hepatocellular toxicity. Toxicol Sci 2024; 201:14-25. [PMID: 38902949 PMCID: PMC11347774 DOI: 10.1093/toxsci/kfae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
Drug-induced liver injury (DILI) is the most common trigger for acute liver failure and the leading cause of attrition in drug development. In this study, we developed an in silico framework to screen drug-induced hepatocellular toxicity (INSIGHT) by integrating the post-treatment transcriptomic data from both rodent models and primary human hepatocytes. We first built an early prediction model using logistic regression with elastic net regularization for 123 compounds and established the INSIGHT framework that can screen for drug-induced hepatotoxicity. The 235 signature genes identified by INSIGHT were involved in metabolism, bile acid synthesis, and stress response pathways. Applying the INSIGHT to an independent transcriptomic dataset treated by 185 compounds predicted that 27 compounds show a high DILI risk, including zoxazolamine and emetine. Further integration with cell image data revealed that predicted compounds with high DILI risk can induce abnormal morphological changes in the endoplasmic reticulum and mitochondrion. Clustering analysis of the treatment-induced transcriptomic changes delineated distinct DILI mechanisms induced by these compounds. Our study presents a computational framework for a mechanistic understanding of long-term liver injury and the prospective prediction of DILI risk.
Collapse
Affiliation(s)
- Yueshan Zhao
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Ji Youn Park
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Da Yang
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, United States
- UPMC Hillman Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, United States
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Min Zhang
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, United States
| |
Collapse
|
9
|
Shen X, Shi C, Xu J, Zhi F, Luo K, Di Y, Li W, Ma W, Jiang Y, Sun H. Intestinal microbiota homeostasis analysis in riboflavin-treated alcoholic liver disease. Commun Biol 2024; 7:1030. [PMID: 39169207 PMCID: PMC11339332 DOI: 10.1038/s42003-024-06722-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Alcoholic liver disease (ALD) is a disease with high incidence, limited therapies, and poor prognosis. The present study aims to investigate the effect of riboflavin on ALD and explore its potential therapeutic mechanisms. C57BL/6 mice were divided into the control, alcohol, and alcohol+ riboflavin groups. 16S rRNA-seq and RNA-seq analysis were utilized to analyze the polymorphism of intestinal microbiota and the transcriptome heterogeneity respectively. KEGG and GO enrichment analysis were performed. CIBERSORTx was applied to evaluate the immune cell infiltration level. Publicly available transcriptome data of ALD was enrolled and combined with the RNA-seq data to identify the immune subtypes of ALD. Pathological and histology analysis demonstrated that riboflavin reversed the progression of ALD. 16S rRNA-seq results showed that riboflavin could regulate alcohol-induced intestinal microbiota alteration. Intestinal microbiota polymorphism analysis indicated that VLIDP may contribute to the progression of ALD. Based on the VLIDP pathway, two subtypes were identified. Immune microenvironment analysis indicated that the upregulated inflammatory factors may be important regulators of ALD. In conclusion, intestinal microbiota homeostasis was associated with the protective effect of riboflavin against ALD, which was likely mediated by modulating inflammatory cell infiltration. Riboflavin emerges as a promising therapeutic candidate for the management of ALD.
Collapse
Affiliation(s)
- Xiuyun Shen
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, PR China
| | - Chunpeng Shi
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Jincheng Xu
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Fengnan Zhi
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Kunpeng Luo
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yuzhu Di
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Wanhong Li
- Pharmaceutical Experiment Teaching Center College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Wanjing Ma
- Pharmaceutical Experiment Teaching Center College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yanan Jiang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, PR China.
| | - Hui Sun
- Pharmaceutical Experiment Teaching Center College of Pharmacy, Harbin Medical University, Harbin, PR China.
| |
Collapse
|
10
|
Nithyasree V, Magdalene P, Praveen Kumar PK, Preethi J, Gromiha MM. Role of HSP90 in Type 2 Diabetes Mellitus and Its Association with Liver Diseases. Mol Biotechnol 2024:10.1007/s12033-024-01251-1. [PMID: 39162909 DOI: 10.1007/s12033-024-01251-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024]
Abstract
Non-alcoholic fatty acid liver disease (NAFLD), non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) are the fatal liver diseases which encompass a spectrum of disease severity associated with increased risk of type 2 diabetes mellitus (T2DM), a metabolic disorder. Heat shock proteins serve as markers in early prognosis and diagnosis of early stages of liver diseases associated with metabolic disorder. This review aims to comprehensively investigate the significance of HSP90 isoforms in T2DM and liver diseases. Additionally, we explore the collective knowledge on plant-based drug compounds that regulate HSP90 isoform targets, highlighting their potential in treating T2DM-associated liver diseases. Furthermore, this review focuses on the computational systems' biology and next-generation sequencing technology approaches that are used to unravel the potential medicine for the treatment of pleiotropy of these 2 diseases and to further elucidate the mechanism.
Collapse
Affiliation(s)
- V Nithyasree
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur Tk, Pennalur, Tamil Nadu, 602117, India
| | - P Magdalene
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur Tk, Pennalur, Tamil Nadu, 602117, India
| | - P K Praveen Kumar
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur Tk, Pennalur, Tamil Nadu, 602117, India.
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India.
| | - J Preethi
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur Tk, Pennalur, Tamil Nadu, 602117, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, 600036, India
| |
Collapse
|
11
|
Kiełbowski K, Skórka P, Plewa P, Bakinowska E, Pawlik A. The Role of Alarmins in the Pathogenesis of Atherosclerosis and Myocardial Infarction. Curr Issues Mol Biol 2024; 46:8995-9015. [PMID: 39194749 DOI: 10.3390/cimb46080532] [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: 06/29/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Atherosclerosis is a condition that is associated with lipid accumulation in the arterial intima. Consequently, the enlarging lesion, which is also known as an atherosclerotic plaque, may close the blood vessel lumen, thus leading to organ ischaemia. Furthermore, the plaque may rupture and initiate the formation of a thrombus, which can cause acute ischaemia. Atherosclerosis is a background pathological condition that can eventually lead to major cardiovascular diseases such as acute coronary syndrome or ischaemic stroke. The disorder is associated with an altered profile of alarmins, stress response molecules that are secreted due to cell injury or death and that induce inflammatory responses. High-mobility group box 1 (HMGB1), S100 proteins, interleukin-33, and heat shock proteins (HSPs) also affect the behaviour of endothelial cells and vascular smooth muscle cells (VSMCs). Thus, alarmins control the inflammatory responses of endothelial cells and proliferation of VSMCs, two important processes implicated in the pathogenesis of atherosclerosis. In this review, we will discuss the role of alarmins in the pathophysiology of atherosclerosis and myocardial infarction.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Patryk Skórka
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Paulina Plewa
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
12
|
Lv Y, Li H, Zhai BT, Sun J, Cheng JX, Zhang XF, Guo DY. Evidence of synergistic mechanisms of hepatoprotective botanical herbal preparation of Pueraria montana var. lobata and Schisandra sphenanthera. Front Pharmacol 2024; 15:1412816. [PMID: 38978983 PMCID: PMC11228302 DOI: 10.3389/fphar.2024.1412816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/03/2024] [Indexed: 07/10/2024] Open
Abstract
Background Pueraria montana var. lobata (Willd.) Maesen & S.M.Almeida ex Sanjappa & Predeep (syn. Pueraria lobata (Willd.) Ohwi) and Schisandra sphenanthera Rehder & E.H. Wilson are traditional edible and medicinal hepatoprotective botanical drugs. Studies have shown that the combination of two botanical drugs enhanced the effects of treating acute liver injury (ALI), but the synergistic effect and its action mechanisms remain unclear. This study aimed to investigate the synergistic effect and its mechanism of the combination of Pueraria montana var. lobata (Willd.) Maesen & S.M.Almeida ex Sanjappa & Predeep (syn. Pueraria lobata (Willd.) Ohwi) (PM) and Schisandra sphenanthera Rehder & E.H. Wilson (SS) in the treatment of ALI. Methods High performance liquid chromatography (HPLC) were utilized to conduct the chemical interaction analysis. Then the synergistic effects of botanical hybrid preparation of PM-SS (BHP PM-SS) against ALI were comprehensively evaluated by the CCl4 induced ALI mice model. Afterwards, symptom-oriented network pharmacology, transcriptomics and metabolomics were applied to reveal the underlying mechanism of action. Finally, the key target genes were experimentally by RT-qPCR. Results Chemical analysis and pharmacodynamic experiments revealed that BHP PM-SS was superior to the single botanical drug, especially at 2:3 ratio, with a better dissolution rate of active ingredients and synergistic anti-ALI effect. Integrated symptom-oriented network pharmacology combined with transcriptomics and metabolomics analyses showed that the active ingredients of BHP PM-SS could regulate Glutathione metabolism, Pyrimidine metabolism, Arginine biosynthesis and Amino acid sugar and nucleotide sugar metabolism, by acting on the targets of AKT1, TNF, EGFR, JUN, HSP90AA1 and STAT3, which could be responsible for the PI3K-AKT signaling pathway, MAPK signaling pathway and Pathway in cancer to against ALI. Conclusion Our study has provided compelling evidence for the synergistic effect and its mechanism of the combination of BHP PM-SS, and has contributed to the development and utilization of BHP PM-SS dietary supplements.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dong-Yan Guo
- State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, College of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, China
| |
Collapse
|
13
|
Roychowdhury S, Pant B, Cross E, Scheraga R, Vachharajani V. Effect of ethanol exposure on innate immune response in sepsis. J Leukoc Biol 2024; 115:1029-1041. [PMID: 38066660 PMCID: PMC11136611 DOI: 10.1093/jleuko/qiad156] [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: 05/10/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
Alcohol use disorder, reported by 1 in 8 critically ill patients, is a risk factor for death in sepsis patients. Sepsis, the leading cause of death, kills over 270,000 patients in the United States alone and remains without targeted therapy. Immune response in sepsis transitions from an early hyperinflammation to persistent inflammation and immunosuppression and multiple organ dysfunction during late sepsis. Innate immunity is the first line of defense against pathogen invasion. Ethanol exposure is known to impair innate and adaptive immune response and bacterial clearance in sepsis patients. Specifically, ethanol exposure is known to modulate every aspect of innate immune response with and without sepsis. Multiple molecular mechanisms are implicated in causing dysregulated immune response in ethanol exposure with sepsis, but targeted treatments have remained elusive. In this article, we outline the effects of ethanol exposure on various innate immune cell types in general and during sepsis.
Collapse
Affiliation(s)
- Sanjoy Roychowdhury
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Bishnu Pant
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Emily Cross
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Rachel Scheraga
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States
| | - Vidula Vachharajani
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States
| |
Collapse
|
14
|
Smith AG, Kliebe VM, Mishra S, McCall RP, Irvine MM, Blagg BSJ, Lei W. Anti-inflammatory activities of novel heat shock protein 90 isoform selective inhibitors in BV-2 microglial cells. Front Mol Biosci 2024; 11:1405339. [PMID: 38756532 PMCID: PMC11096514 DOI: 10.3389/fmolb.2024.1405339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Heat shock protein 90 (Hsp90) is a family of chaperone proteins that consists of four isoforms: Hsp90α, Hsp90β, glucose-regulated protein 94 (Grp94), and tumor necrosis factor type 1 receptor-associated protein (TRAP1). They are involved in modulating the folding, maturation, and activation of their client proteins to regulate numerous intracellular signaling pathways. Previous studies demonstrated that pan-Hsp90 inhibitors reduce inflammatory signaling pathways resulting in a reduction of inflammation and pain but show toxicities in cancer-related clinical trials. Further, the role of Hsp90 isoforms in inflammation remains poorly understood. This study aimed to determine anti-inflammatory activities of Hsp90 isoforms selective inhibitors on the lipopolysaccharide (LPS)-induced inflammation in BV-2 cells, a murine microglial cell line. The production of inflammatory mediators such as nitric oxide (NO), interleukin 1 beta (IL-1β), and tumor necrosis factor-alpha (TNF-α) was measured. We also investigated the impact of Hsp90 isoform inhibitors on the activation of nuclear factor kappa B (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), and mitogen-activated protein kinases (MAPKs). We found that selective inhibitors of Hsp90β reduced the LPS-induced production of NO, IL-1β, and TNF-α via diminishing the activation of NF-κB and Extracellular signal-regulated kinases (ERK) MAPK. The Hsp90α, Grp94, TRAP1 inhibitors had limited effect on the production of inflammatory mediators. These findings suggest that Hsp90β is the key player in LPS-induced neuroinflammation. Thereby providing a more selective drug target for development of medications involved in pain management that can potentially contribute to the reduction of adverse side effects associated with Hsp90 pan inhibitors.
Collapse
Affiliation(s)
- Amanda G. Smith
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC, United States
| | | | - Sanket Mishra
- Department of Chemistry and Biochemistry, University of Notre Dame College of Science, Notre Dame, IN, United States
| | - Ryan P. McCall
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC, United States
| | - Megan M. Irvine
- Department of Pharmaceutical and Graduate Life Sciences, Manchester University Fort Wayne, Fort Wayne, IN, United States
| | - Brian S. J. Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame College of Science, Notre Dame, IN, United States
| | - Wei Lei
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC, United States
- Department of Pharmaceutical and Graduate Life Sciences, Manchester University Fort Wayne, Fort Wayne, IN, United States
| |
Collapse
|
15
|
Sayaf K, Battistella S, Russo FP. NLRP3 Inflammasome in Acute and Chronic Liver Diseases. Int J Mol Sci 2024; 25:4537. [PMID: 38674122 PMCID: PMC11049922 DOI: 10.3390/ijms25084537] [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: 01/29/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) is an intracellular complex that upon external stimuli or contact with specific ligands, recruits other components, forming the NLRP3 inflammasome. The NLRP3 inflammasome mainly mediates pyroptosis, a highly inflammatory mode of regulated cell death, as well as IL-18 and IL-1β production. Acute and chronic liver diseases are characterized by a massive influx of pro-inflammatory stimuli enriched in reactive oxygen species (ROS) and damage-associated molecular patterns (DAMPs) that promote the assemblage and activation of the NLRP3 inflammasome. As the major cause of inflammatory cytokine storm, the NLRP3 inflammasome exacerbates liver diseases, even though it might exert protective effects in regards to hepatitis C and B virus infection (HCV and HBV). Here, we summarize the current knowledge concerning NLRP3 inflammasome function in both acute and chronic liver disease and in the post liver transplant setting, focusing on the molecular mechanisms involved in NLRP3 activity.
Collapse
Affiliation(s)
- Katia Sayaf
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy; (K.S.); (S.B.)
| | - Sara Battistella
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy; (K.S.); (S.B.)
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, 35128 Padua, Italy
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy; (K.S.); (S.B.)
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, 35128 Padua, Italy
| |
Collapse
|
16
|
She Y, Guo Z, Zhai Q, Liu J, Du Q, Zhang Z. CDK4/6 inhibitors in drug-induced liver injury: a pharmacovigilance study of the FAERS database and analysis of the drug-gene interaction network. Front Pharmacol 2024; 15:1378090. [PMID: 38633610 PMCID: PMC11021785 DOI: 10.3389/fphar.2024.1378090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/20/2024] [Indexed: 04/19/2024] Open
Abstract
Objective The aim of this study was to investigate the potential risk of drug-induced liver injury (DILI) caused by the CDK4/6 inhibitors (CDK4/6is abemaciclib, ribociclib, and palbociclib by comprehensively analyzing the FDA Adverse Event Reporting System (FAERS) database. Moreover, potential toxicological mechanisms of CDK4/6is-related liver injury were explored via drug-gene network analysis. Methods In this retrospective observational study, we collected reports of DILI associated with CDK4/6i use from the FAERS dated January 2014 to March 2023. We conducted disproportionality analyses using the reporting odds ratio (ROR) with a 95% confidence interval (CI). Pathway enrichment analysis and drug-gene network analyses were subsequently performed to determine the potential mechanisms underlying CDK4/6i-induced liver injury. Results We found positive signals for DILI with ribociclib (ROR = 2.60) and abemaciclib (ROR = 2.37). DILIs associated with liver-related investigations, signs, and symptoms were confirmed in all three reports of CDK4/6is. Moreover, ascites was identified as an unlisted hepatic adverse effect of palbociclib. We isolated 189 interactive target genes linking CDK4/6 inhibitors to hepatic injury. Several key genes, such as STAT3, HSP90AA1, and EP300, were revealed via protein-protein analysis, emphasizing their central roles within the network. KEGG pathway enrichment of these genes highlighted multiple pathways. Conclusion Our study revealed variations in hepatobiliary toxicity among the different CDK4/6 inhibitors, with ribociclib showing the highest risk of liver injury, followed by abemaciclib, while palbociclib appeared relatively safe. Our findings emphasize the need for cautious use of CDK4/6 inhibitors, and regular liver function monitoring is recommended for long-term CDK4/6 inhibitor use.
Collapse
Affiliation(s)
- Youjun She
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zihan Guo
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Zhai
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiong Du
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongwei Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Critical Care, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
17
|
Huang J, Huang T, Li J. Regulation Mechanism and Potential Value of Active Substances in Spices in Alcohol-Liver-Intestine Axis Health. Int J Mol Sci 2024; 25:3728. [PMID: 38612538 PMCID: PMC11011869 DOI: 10.3390/ijms25073728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Excessive alcohol intake will aggravate the health risk between the liver and intestine and affect the multi-directional information exchange of metabolites between host cells and microbial communities. Because of the side effects of clinical drugs, people tend to explore the intervention value of natural drugs on diseases. As a flavor substance, spices have been proven to have medicinal value, but they are still rare in treating hepatointestinal diseases caused by alcohol. This paper summarized the metabolic transformation of alcohol in the liver and intestine and summarized the potential value of various perfume active substances in improving liver and intestine diseases caused by alcohol. It is also found that bioactive substances in spices can exert antioxidant activity in the liver and intestine environment and reduce the oxidative stress caused by diseases. These substances can interfere with fatty acid synthesis, promote sugar and lipid metabolism, and reduce liver injury caused by steatosis. They can effectively regulate the balance of intestinal flora, promote the production of SCFAs, and restore the intestinal microenvironment.
Collapse
Affiliation(s)
- Jianyu Huang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China;
- College of Food and Pharmaceutical Science, Ningbo University, Ningbo 315211, China
| | - Tao Huang
- College of Food and Pharmaceutical Science, Ningbo University, Ningbo 315211, China
| | - Jinjun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China;
| |
Collapse
|
18
|
Dederichs TS, Yerdenova A, Horstmann H, Vico TA, Nübling S, Peyronnet R, Pfeifer D, von zur Muehlen C, Heidt T, Wolf D, Czerny M, Westermann D, Hilgendorf I. Nonpreferential but Detrimental Accumulation of Macrophages With Clonal Hematopoiesis-Driver Mutations in Cardiovascular Tissues-Brief Report. Arterioscler Thromb Vasc Biol 2024; 44:690-697. [PMID: 38269586 PMCID: PMC10880934 DOI: 10.1161/atvbaha.123.320183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/04/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP) is an acquired genetic risk factor for both leukemia and cardiovascular disease. It results in proinflammatory myeloid cells in the bone marrow and blood; however, how these cells behave in the cardiovascular tissue remains unclear. Our study aimed at investigating whether CHIP-mutated macrophages accumulate preferentially in cardiovascular tissues and examining the transcriptome of tissue macrophages from DNMT3A (DNA methyltransferase 3 alpha) or TET2 (Tet methylcytosine dioxygenase 2) mutation carriers. METHODS We recruited patients undergoing carotid endarterectomy or heart surgeries to screen for CHIP mutation carriers using targeted genomic sequencing. Myeloid and lymphoid cells were isolated from blood and cardiovascular tissue collected during surgeries using flow cytometry. DNA and RNA extracted from these sorted cells were subjected to variant allele frequency measurement using droplet digital polymerase chain reaction and transcriptomic profiling using bulk RNA sequencing, respectively. RESULTS Using droplet digital polymerase chain reaction, we detected similar variant allele frequency of CHIP in monocytes from blood and macrophages from atheromas and heart tissues, even among heart macrophages with and without CCR2 (C-C motif chemokine receptor 2) expression. Bulk RNA sequencing revealed a proinflammatory gene profile of myeloid cells from DNMT3A or TET2 mutation carriers compared with those from noncarriers. CONCLUSIONS Quantitatively, CHIP-mutated myeloid cells did not preferentially accumulate in cardiovascular tissues, but qualitatively, they expressed a more disease-prone phenotype.
Collapse
Affiliation(s)
- Tsai-Sang Dederichs
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
| | - Assel Yerdenova
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
| | - Hauke Horstmann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
| | - Tamara Antonela Vico
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
| | - Simone Nübling
- Institute for Experimental Cardiovascular Medicine, Cardio-Vascular Biobank, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (S.N., R.P., I.H.)
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, Cardio-Vascular Biobank, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (S.N., R.P., I.H.)
| | - Dietmar Pfeifer
- Department of Internal Medicine I, Medical Center and Faculty of Medicine, University of Freiburg, Germany (D.P.)
| | - Constantin von zur Muehlen
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
| | - Timo Heidt
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine University of Freiburg, Germany (M.C.)
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (T.-S.D., A.Y., H.H., T.A.V., C.v.z.M., T.H., D. Wolf, D. Westermann, I.H.)
- Institute for Experimental Cardiovascular Medicine, Cardio-Vascular Biobank, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine, University of Freiburg, Germany (S.N., R.P., I.H.)
| |
Collapse
|
19
|
Wang J, Wu Z, Chen X, Sun Y, Ma S, Weng J, Zhang Y, Dong K, Shao J, Zheng S. Network Pharmacology, Molecular Docking Analysis and Molecular Dynamics Simulation of Scutellaria baicalensis in the Treatment of Liver Fibrosis. Curr Pharm Des 2024; 30:1326-1340. [PMID: 38616754 DOI: 10.2174/0113816128297074240327090020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Traditional Chinese medicine Scutellaria Baicalensis (SB), one of the clinical firstline heat-clearing drugs, has obvious symptomatic advantages for hepatic fibrosis with dampness-heat stasis as its syndrome. We aim to predict and validate the potential mechanism of Scutellaria baicalensis active ingredients against liver fibrosis more scientifically and effectively. METHODS The underlying mechanism of Scutellaria baicalensis in inhibiting hepatic fibrosis was studied by applying network pharmacology, molecular docking and molecular dynamics simulation. Expression levels of markers in activated Hepatic Stellate Cells (HSC) after administration of three Scutellaria baicalensis extracts were determined by Western blot and Real-time PCR, respectively, in order to verify the anti-fibrosis effect of the active ingredients Results: There are 164 common targets of drugs and diseases screened and 115 signaling pathways obtained, which were mainly associated with protein phosphorylation, senescence and negative regulation of the apoptotic process. Western blot and Real-time PCR showed that Scutellaria baicalensis extracts could reduce the expression of HSC activation markers, and Oroxylin A had the strongest inhibitory effect on it. Molecular docking results showed that Oroxylin A had high binding activity to target proteins. Molecular dynamics simulation demonstrates promising stability of the Oroxylin A-AKT1 complex over the simulated MD time of 200 ns. CONCLUSION Scutellaria baicalensis active ingredients may inhibit HSC proliferation, reduce the generation of pro-inflammatory factors and block the anti-inflammatory effect of inflammatory signal transduction by inducing HSC apoptosis and senescence, thus achieving the effect of anti-fibrosis.
Collapse
Affiliation(s)
- Junrui Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhuoqing Wu
- Nanjing Foreign Language School, Nanjing, China
| | - Xiaolei Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Sun
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuyao Ma
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingdan Weng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Keke Dong
- PharmaBlock Sciences (Nanjing), Inc, Nanjing, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Sklifasovskaya AP, Blagonravov M, Ryabinina A, Goryachev V, Syatkin S, Chibisov S, Akhmetova K, Prokofiev D, Agostinelli E. The role of heat shock proteins in the pathogenesis of heart failure (Review). Int J Mol Med 2023; 52:106. [PMID: 37772383 PMCID: PMC10558216 DOI: 10.3892/ijmm.2023.5309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/11/2023] [Indexed: 09/30/2023] Open
Abstract
The influence of heat shock proteins (HSPs) on protein quality control systems in cardiomyocytes is currently under investigation. The effect of HSPs on the regulated cell death of cardiomyocytes (CMCs) is of great importance, since they play a major role in the implementation of compensatory and adaptive mechanisms in the event of cardiac damage. HSPs mediate a number of mechanisms that activate the apoptotic cascade, playing both pro‑ and anti‑apoptotic roles depending on their location in the cell. Another type of cell death, autophagy, can in some cases lead to cell death, while in other situations it acts as a cell survival mechanism. The present review considered the characteristics of the expression of HSPs of different molecular weights in CMCs in myocardial damage caused by heart failure, as well as their role in the realization of certain types of regulated cell death.
Collapse
Affiliation(s)
| | | | - Anna Ryabinina
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | | | - Sergey Syatkin
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Sergey Chibisov
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Karina Akhmetova
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Daniil Prokofiev
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Faculty of Medicine and Dentistry, Sapienza University of Rome, University Hospital Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation, ETS-ONLUS, I-00159 Rome, Italy
| |
Collapse
|
21
|
Stanca L, Geicu OI, Serban AI, Dinischiotu A. Interplay of Oxidative Stress, Inflammation, and Autophagy in RAW 264.7 Murine Macrophage Cell Line Challenged with Si/SiO 2 Quantum Dots. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5083. [PMID: 37512357 PMCID: PMC10385521 DOI: 10.3390/ma16145083] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/07/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023]
Abstract
Quantum dots (QDs) with photostable fluorescence are recommended for imaging applications; however, their effect on living cells is incompletely understood. We aimed to elucidate the RAW 264.7 murine macrophage cell line's response to the Si/SiO2 QDs challenge. Cells were exposed to 5 and 15 μg/mL Si/SiO2 QDs for 6 h, 12 h, and 24 h. Cell metabolic activity and viability were assessed by MTT, live/dead, and dye-exclusion assays. Oxidative stress and membrane integrity were assessed by anion superoxide, malondialdehyde, and lactate dehydrogenase activity evaluations. Antioxidative enzyme activities were analyzed by kinetic spectrophotometric methods. Cytokines were analyzed with an antibody-based magnetic bead assay, PGE2 was assessed by ELISA, and Nrf-2, Bcl-2, Beclin 1, and the HSPs were analyzed by western blot. Autophagy levels were highlighted by fluorescence microscopy. The average IC50 dose for 6, 12, and 24 h was 16.1 ± 0.7 μg/mL. Although glutathione S-transferase and catalase were still upregulated after 24 h, superoxide dismutase was inhibited, which together allowed the gradual increase of malondialdehyde, anion superoxide, nitric oxide, and the loss of membrane integrity. G-CSF, IL-6, TNF-α, MIP-1β, MCP-1, Nrf-2, PGE2, and RANTES levels, as well as autophagy processes, were increased at all time intervals, as opposed to caspase 1 activity, COX-2, HSP60, and HSP70, which were only upregulated at the 6-h exposure interval. These results underscore that Si/SiO2 QDs possess significant immunotoxic effects on the RAW 264.7 macrophage cell line and stress the importance of developing effective strategies to mitigate their adverse impact.
Collapse
Affiliation(s)
- Loredana Stanca
- Preclinical Sciences Department, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, 050097 Bucharest, Romania
| | - Ovidiu Ionut Geicu
- Preclinical Sciences Department, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, 050097 Bucharest, Romania
| | - Andreea Iren Serban
- Preclinical Sciences Department, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, 050097 Bucharest, Romania
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| |
Collapse
|
22
|
Qiu T, Shi JX, Cheng C, Jiang H, Ruan HN, Li J, Liu CM. Hepatoprotective effect of avicularin on lead-induced steatosis, oxidative stress, and inflammation in mice associated with the MAPK/HSP60/NLRP3 and SREBP1c pathway. Toxicol Res (Camb) 2023; 12:417-424. [PMID: 37397929 PMCID: PMC10311149 DOI: 10.1093/toxres/tfad028] [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: 11/07/2022] [Revised: 02/20/2023] [Accepted: 03/31/2023] [Indexed: 07/04/2023] Open
Abstract
Lead (Pb), an environmental hazard, causes severe diseases in the liver, kidney, cardiovascular system, hematopoietic system, reproductive system, and nervous system. Avicularin (AVI), the main dietary flavonoid found in many citrus fruits, exhibited potential protective properties on organs. However, the molecular mechanisms of these protective actions are currently not clear. In our study, the effects of AVI on Pb-induced hepatotoxicity were evaluated using ICR mice. Changes in oxidative stress, inflammation, lipid metabolism, and related signaling were evaluated. We found for the first time that treatment with AVI significantly reduced hepatic steatosis, inflammation, and oxidative stress induced by Pb. AVI attenuated Pb-induced liver dysfunction and lipid metabolism disorder in mice. AVI decreased the serum biochemical indicators of lipid metabolism. AVI decreased the expression levels of lipid metabolism-related protein SREBP-1c, acetyl-CoA carboxylase (ACC), and FAS. AVI suppressed Pb-induced inflammation in livers, as indicated by decreasing the TNF-α and IL-1β levels. AVI suppressed oxidative stress by increasing the activation of SOD, CAT, and GPx. Furthermore, AVI inhibited the activities of JNK, ERK, p38, and NF-κB. AVI further decreased the levels of HSP60, NLRP3, p-IκBα, and p-p65 in the livers of mice. Collectively, this study indicated that AVI mitigated Pb-induced hepatic steatosis, oxidative stress, and inflammation by regulating the SREBP-1c and MAPK/HSP60/NLRP3 signaling pathways.
Collapse
Affiliation(s)
- Ting Qiu
- School of Life Science, Jiangsu Normal University, No.101, Shanghai Road, Tongshan New Area, Xuzhou City, Jiangsu Province 21-1116, PR China
| | - Jia-Xue Shi
- School of Life Science, Jiangsu Normal University, No.101, Shanghai Road, Tongshan New Area, Xuzhou City, Jiangsu Province 21-1116, PR China
| | - Chao Cheng
- School of Life Science, Jiangsu Normal University, No.101, Shanghai Road, Tongshan New Area, Xuzhou City, Jiangsu Province 21-1116, PR China
| | - Hong Jiang
- School of Life Science, Jiangsu Normal University, No.101, Shanghai Road, Tongshan New Area, Xuzhou City, Jiangsu Province 21-1116, PR China
| | - Hai-Nan Ruan
- School of Life Science, Jiangsu Normal University, No.101, Shanghai Road, Tongshan New Area, Xuzhou City, Jiangsu Province 21-1116, PR China
| | - Jun Li
- School of Life Science, Jiangsu Normal University, No.101, Shanghai Road, Tongshan New Area, Xuzhou City, Jiangsu Province 21-1116, PR China
| | - Chan-Min Liu
- School of Life Science, Jiangsu Normal University, No.101, Shanghai Road, Tongshan New Area, Xuzhou City, Jiangsu Province 21-1116, PR China
| |
Collapse
|
23
|
Ma J, Li M, Yang T, Deng Y, Ding Y, Guo T, Shang J. Isoquercitrin Attenuates Steatohepatitis by Inhibition of the Activated NLRP3 Inflammasome through HSP90. Int J Mol Sci 2023; 24:ijms24108795. [PMID: 37240141 DOI: 10.3390/ijms24108795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/10/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease with a global prevalence of 25%. However, the medicines approved by the FDA or EMA are still not commercially available for the treatment of NAFLD. The NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome plays a crucial role in inflammatory responses, and the mechanisms related to steatohepatitis have been sufficiently clarified. NLRP3 has been widely evaluated as a potential target for multiple active agents in treating NAFLD. As a quercetin glycoside, isoquercitrin (IQ) has a broad inhibitory effect on oxidative stress, cancers, cardiovascular diseases, diabetes, and allergic reactions in vitro and in vivo. This study aimed to investigate the undercover mechanism of IQ in the treatment of NAFLD, particularly in anti-steatohepatitis, by suppressing the NLRP3 inflammasome. In this study, a methionine-choline-deficient induced steatohepatitis mice model was used to explore the effect of IQ on NAFLD treatment. Further mechanism exploration based on transcriptomics and molecular biology revealed that IQ inhibited the activated NLRP3 inflammasome by down-regulating the expression of heat shock protein 90 (HSP90) and suppressor of G-two allele of Skp1 (SGT1). In conclusion, IQ could alleviate NAFLD by inhibiting the activated NLRP3 inflammasome by suppressing the expression of HSP90.
Collapse
Affiliation(s)
- Ji Ma
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China
| | - Maoru Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China
| | - Tingting Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Deng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yadong Ding
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China
| | - Tiantian Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China
| | - Jing Shang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
24
|
Tan Y, Zhang F, Fan X, Lu S, Liu Y, Wu Z, Huang Z, Wu C, Cheng G, Li B, Huang J, Stalin A, Zhou W, Wu J. Exploring the effect of Yinzhihuang granules on alcoholic liver disease based on pharmacodynamics, network pharmacology and molecular docking. Chin Med 2023; 18:52. [PMID: 37165407 PMCID: PMC10173499 DOI: 10.1186/s13020-023-00759-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Yinzhihuang granules (YZHG) is a commonly used Chinese patent medicine for the treatment of liver disease. However, the mechanism of YZHG in alcoholic liver disease (ALD) is still unclear. METHODS This study combined liquid chromatography-mass spectrometry technology, pharmacodynamics, network pharmacology and molecular docking methods to evaluate the potential mechanism of YZHG in the treatment of ALD. RESULTS A total of 25 compounds including 4-hydroxyacetophenone, scoparone, geniposide, quercetin, baicalin, baicalein, chlorogenic acid and caffeic acid in YZHG were identified by ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). The pharmacodynamic investigations indicated that YZHG could improve liver function and the degree of liver tissue lesions, and reduce liver inflammation and oxidative stress in ALD mice. Network pharmacology analysis showed that YZHG treated ALD mainly by regulating inflammation-related signaling pathways such as the PI3K-Akt signaling pathway. The results of the PPI network and molecular docking showed that the targets of SRC, HSP90AA1, STAT3, EGFR and AKT1 could be the key targets of YZHG in the treatment of ALD. CONCLUSION This study explored the potential compounds, potential targets and signaling pathways of YZHG in the treatment of ALD, which is helpful to clarify the efficacy and mechanism of YZHG and provide new insights for the clinical application of YZHG.
Collapse
Affiliation(s)
- Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fanqin Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotian Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guoliang Cheng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Bing Li
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jiaqi Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.
| | - Wei Zhou
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
25
|
Xiang W, Yang Y, Weng L, Ye Z, Ding P, Li H, Sun J, Zeng C. Hyperhomocysteinemia activates NLRP3 inflammasome to cause hepatic steatosis and insulin resistance via MDM2-mediated ubiquitination of HSF1. Int Immunopharmacol 2023; 118:110085. [PMID: 37018978 DOI: 10.1016/j.intimp.2023.110085] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Hyperhomocysteinemia (HHcy) is associated with nonalcoholic fatty liver disease (NAFLD) and insulin resistance (IR). However, the underlying mechanism is still unknown. Recent studies have demonstrated that NLRP3 inflammasome activation plays a vital role in NAFLD and IR. Our study aimed to explore whether NLRP3 inflammasome contributed to HHcy-induced NAFLD and IR as well as dissected the underlying mechanism. C57BL/6 mice were fed a high-methionine diet (HMD) for 8 weeks to establish the HHcy mouse model. Compared with a chow diet, HMD induced hepatic steatosis (HS) and IR as well as activation of hepatic NLRP3 inflammasome. Moreover, HHcy-induced NAFLD and IR characterization disclosed that NLRP3 inflammasome activation occurred in liver tissue of HMD-fed mice, but was very marginal in either NLRP3-/- or Caspase-1-/- mice. Mechanistically, high levels of homocysteine (Hcy) up-regulated the expression of mouse double minute 2 homolog (MDM2), which directly ubiquitinates heat shock transcription factor 1 (HSF1) and consequently activated hepatic NLRP3 inflammasome in vivo and in vitro. In addition, in vitro experiments showed P300-mediated HSF1 acetylation at K298 hindered MDM2-mediated ubiquitination of HSF1 at K372, which plays important role in determining the HSF1 level. Importantly, either inhibition of MDM2 by JNJ-165 or activation of HSF1 by HSF1A reversed HMD-induced hepatic NLRP3 inflammasome, and consequently alleviated HS and IR in mice. This study demonstrates that NLRP3 inflammasome activation contributes to HHcy-induced NAFLD and IR, and further identified that HSF1 as a new substrate of MDM2 and its decrease on MDM2-mediated ubiquitination at K372 modulates NLRP3 inflammasome activation. These findings may provide novel therapeutic strategies aimed at halting HS or IR.
Collapse
Affiliation(s)
- Wenjing Xiang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yang Yang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, China
| | - Liangkun Weng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhiming Ye
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ping Ding
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huayu Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jia Sun
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Cheng Zeng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, 510699, China; Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
26
|
Adjei‐Mosi J, Sun Q, Smithson SB, Shealy GL, Amerineni KD, Liang Z, Chen H, Wang M, Ping Q, Han J, Morita M, Kamat A, Musi N, Zang M. Age-dependent loss of hepatic SIRT1 enhances NLRP3 inflammasome signaling and impairs capacity for liver fibrosis resolution. Aging Cell 2023; 22:e13811. [PMID: 36999514 PMCID: PMC10186605 DOI: 10.1111/acel.13811] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 04/01/2023] Open
Abstract
Our studies indicate that the longevity factor SIRT1 is implicated in metabolic disease; however, whether and how hepatocyte-specific SIRT1 signaling is involved in liver fibrosis remains undefined. We characterized a functional link of age-mediated defects in SIRT1 to the NLRP3 inflammasome during age-related liver fibrosis. In multiple experimental murine models of liver fibrosis, we compared the development of liver fibrosis in young and old mice, as well as in liver-specific SIRT1 knockout (SIRT1 LKO) mice and wild-type (WT) mice. Liver injury, fibrosis, and inflammation were assessed histologically and quantified by real-time PCR analysis. In a model of hepatotoxin-induced liver fibrosis, old mice displayed more severe and persistent liver fibrosis than young mice during liver injury and after injury cessation, as characterized by inhibition of SIRT1, induction of NLRP3, infiltration of macrophages and neutrophils, activation of hepatic stellate cells (HSCs), and excessive deposition and remodeling of the extracellular matrix. Mechanistically, deletion of SIRT1 in hepatocytes resulted in NLRP3 and IL-1β induction, pro-inflammatory response, and severe liver fibrosis in young mice, mimicking the ability of aging to impair the resolution of established fibrosis. In an aging mouse model, chronic-plus-binge alcohol feeding-induced liver fibrosis was attenuated by treatment with MCC950, a selective NLRP3 inhibitor. NLRP3 inhibition ameliorated alcoholic liver fibrosis in old mice by repressing inflammation and reducing hepatocyte-derived danger signaling-ASK1 and HMGB1. In conclusion, age-dependent SIRT1 defects lead to NLRP3 activation and inflammation, which in turn impairs the capacity to resolve fibrosis during aging.
Collapse
Affiliation(s)
- Jennifer Adjei‐Mosi
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Qing Sun
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Steven Blake Smithson
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Gavyn Lee Shealy
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Krupa Dhruvitha Amerineni
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Zerong Liang
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Hanqing Chen
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Mei Wang
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Qinggong Ping
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Jingyan Han
- Boston University School of MedicineBostonMassachusettsUSA
| | - Masahiro Morita
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
| | - Amrita Kamat
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Geriatric Research, Education and Clinical CenterSouth Texas Veterans Health Care SystemSan AntonioTexasUSA
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Geriatric Research, Education and Clinical CenterSouth Texas Veterans Health Care SystemSan AntonioTexasUSA
| | - Mengwei Zang
- Barshop Institute for Longevity and Aging Studies, Center for Healthy AgingSan AntonioTexasUSA
- Department of Molecular MedicineThe University of Texas Health San AntonioSan AntonioTexasUSA
- Geriatric Research, Education and Clinical CenterSouth Texas Veterans Health Care SystemSan AntonioTexasUSA
| |
Collapse
|
27
|
HSF1 Attenuates the Release of Inflammatory Cytokines Induced by Lipopolysaccharide through Transcriptional Regulation of Atg10. Microbiol Spectr 2023; 11:e0305922. [PMID: 36598250 PMCID: PMC9927406 DOI: 10.1128/spectrum.03059-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Autophagy plays an important role in endotoxemic mice, and heat shock factor 1 (HSF1) plays a crucial protective role in endotoxemic mice. However, the protective mechanisms of HSF1 are poorly understood. In this text, bioinformatics analysis, chromatin immunoprecipitation, and electrophoresis mobility shift assay were employed to investigate the underlying mechanisms. The results showed that the release of inflammatory cytokines increased and autophagy decreased significantly in Hsf1-/- endotoxemic mice compared with those in Hsf1+/+ endotoxemic mice. HSF1 could directly bind to the noncoding promoter region of the autophagy-related gene 10 (Atg10). The expression of ATG10 and the ratio of LC3-II/LC3-I were obviously decreased in LPS-treated Hsf1-/- peritoneal macrophages (PM) versus those in LPS-treated Hsf1+/+ PM. Overexpression of HSF1 increased the level of the ATG10 protein and enhanced the ratio of LC3-II/LC3-I in RAW264.7 cells. In contrast, silencing of HSF1 decreased the expression of ATG10 and markedly lowered the ratio of LC3-II/LC3-I. In a cotransfected cell experiment, the upregulation of autophagy by overexpression HSF1 was reversed by small interfering RNA (siRNA)-ATG10. Compared with the overexpression HSF1, the release of inflammatory cytokines induced by lipopolysaccharide (LPS) was decreased in pcDNA3.1-HSF1 with siRNA-ATG10 cotransfected RAW264.7 cells. On the other hand, the decrease of autophagy by siRNA-HSF1 was compensated by overexpression of ATG10. Compared with siRNA-HSF1, the release of inflammatory cytokines induced by LPS was increased in siRNA-HSF1 with pcDNA3.1-ATG10 cotransfected RAW264.7 cells. These results presented a novel mechanism that HSF1 attenuated the release of inflammatory cytokines induced by LPS through transcriptional regulation of Atg10. Targeting of HSF1-Atg10-autophagy might be an attractive strategy in endotoxemia therapeutics. IMPORTANCE HSF1 plays an important protective role in endotoxemic mice. However, the protective mechanisms of HSF1 are poorly understood. In the present study, we demonstrated that HSF1 upregulated ATG10 through specifically binding Atg10 promoter's noncoding region in LPS-treated PM and RAW264.7 cells. By depletion of HSF1, the expression of ATG10 was significantly decreased, leading to aggravate releasing of inflammatory cytokines in LPS-treated RAW264.7 cells. These findings provided a new mechanism of HSF1 in endotoxemic mice.
Collapse
|
28
|
He YF, Hu XM, Khan MA, Yu BY, Sheng YC, Xiao XZ, Wan XX, Tan SP, Xiong K. HSF1 Alleviates Brain Injury by Inhibiting NLRP3-Induced Pyroptosis in a Sepsis Model. Mediators Inflamm 2023; 2023:2252255. [PMID: 36741074 PMCID: PMC9897924 DOI: 10.1155/2023/2252255] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/15/2022] [Accepted: 09/16/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Sepsis, which could cause a systemic inflammatory response, is a life-threatening disease with a high morbidity and mortality rate. There is evidence that brain injury may be related to severe systemic infection induced by sepsis. The brain injury caused by sepsis could increase the risk of mortality in septic patients, which seriously affects the septic patient's prognosis of survival. Although there remains a focus on sepsis research, clinical measures to prevent and treat brain injury in sepsis are not yet available, and the high mortality rate is still a big health burden. Therefore, it is necessary to investigate the new molecules or regulated pathways that can effectively inhibit the progress of sepsis. OBJECTIVE NLR family pyrin domain-containing 3 (NLRP3) increased in the procession of sepsis and functioned as the key regulator of pyroptosis. Heat shock factor 1 (HSF1) can protect organs from multiorgan dysfunction syndrome induced by lipopolysaccharides in mice, and NLRP3 could be inhibited by HSF1 in many organs. However, whether HSF1 regulated NLRP3 in sepsis-induced brain injury, as well as the detailed mechanism of HSF1 in brain injury, remains unknown in the sepsis model. In this research, we try to explore the relationship between HSF1 and NLRP3 in a sepsis model and try to reveal the mechanism of HSF1 inhibiting the process of brain injury. METHODS In this study, we used wild-type mice and hsf1 -/- mice for in vivo research and PC12 cells for in vitro research. Real-time PCR and Western blot were used to analyze the expression of HSF1, NLRP3, cytokines, and pyrolytic proteins. EthD-III staining was chosen to detect the pyroptosis of the hippocampus and PC12 cells. RESULTS The results showed that HSF1 is negatively related to pyroptosis. The pyroptosis in cells of brain tissue was significantly increased in the hsf1 -/- mouse model compared to hsf1 +/+ mice. In PC12 cells, hsf1 siRNA can upregulate pyroptosis while HSF1-transfected plasmid could inhibit the pyroptosis. HSF1 could negatively regulate the NLRP3 pathway in PC12 cells, while hsf1 siRNA enhanced the pyroptosis in PC12 cells, which could be reversed by nlrp3 siRNA. CONCLUSION These results imply that HSF1 could alleviate sepsis-induced brain injury by inhibiting pyroptosis through the NLRP3-dependent pathway in brain tissue and PC12 cells, suggesting HSF1 as a potential molecular target for treating brain injury in sepsis clinical studies.
Collapse
Affiliation(s)
- Yi-fu He
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xi-min Hu
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Md. Asaduzzaman Khan
- The Research Centre for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Bo-yao Yu
- Clinical Medicine Five-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Yi-cun Sheng
- Clinical Medicine Five-Year Program, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Xian-zhong Xiao
- Key Laboratory of Sepsis Translational Medicine of Hunan, Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Xin-xing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Si-pin Tan
- Key Laboratory of Sepsis Translational Medicine of Hunan, Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Ophthalmology, Changsha 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
29
|
Crotty K, Anton P, Coleman LG, Morris NL, Lewis SA, Samuelson DR, McMahan RH, Hartmann P, Kim A, Ratna A, Mandrekar P, Wyatt TA, Choudhry MA, Kovacs EJ, McCullough R, Yeligar SM. A critical review of recent knowledge of alcohol's effects on the immunological response in different tissues. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:36-44. [PMID: 36446606 PMCID: PMC9974783 DOI: 10.1111/acer.14979] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
Abstract
Alcohol misuse contributes to the dysregulation of immune responses and multiorgan dysfunction across various tissues, which are associated with higher risk of morbidity and mortality in people with alcohol use disorders. Organ-specific immune cells, including microglia in the brain, alveolar macrophages in the lungs, and Kupffer cells in the liver, play vital functions in host immune defense through tissue repair and maintenance of homeostasis. However, binge drinking and chronic alcohol misuse impair these immune cells' abilities to regulate inflammatory signaling and metabolism, thus contributing to multiorgan dysfunction. Further complicating these delicate systems, immune cell dysfunction associated with alcohol misuse is exacerbated by aging and gut barrier leakage. This critical review describes recent advances in elucidating the potential mechanisms by which alcohol misuse leads to derangements in host immunity and highlights current gaps in knowledge that may be the focus of future investigations.
Collapse
Affiliation(s)
- Kathryn Crotty
- Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Paige Anton
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
- Alcohol Research Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Leon G Coleman
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Niya L Morris
- Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Sloan A Lewis
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Derrick R Samuelson
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Rachel H McMahan
- Alcohol Research Program, University of Colorado Denver, Aurora, Colorado, USA
- Department of Surgery, University of Colorado, Aurora, Colorado, USA
| | - Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Adam Kim
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Anuradha Ratna
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Pranoti Mandrekar
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Todd A Wyatt
- Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Mashkoor A Choudhry
- Alcohol Research Program, Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, Maywood, Illinois, USA
| | - Elizabeth J Kovacs
- Alcohol Research Program, University of Colorado Denver, Aurora, Colorado, USA
- Department of Surgery, University of Colorado, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs (VA) Medical Center, Aurora, Colorado, USA
| | - Rebecca McCullough
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
- Alcohol Research Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Samantha M Yeligar
- Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| |
Collapse
|
30
|
Chen L, Fan H, Chu H, Du F, Chen Y, Hu L, Li Z, Wang W, Hou X, Yang L. The HSP90 inhibitor 17-DMAG alleviates primary biliary cholangitis via cholangiocyte necroptosis prevention. J Cell Biochem 2022; 123:1857-1872. [PMID: 36037317 DOI: 10.1002/jcb.30321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/07/2022] [Accepted: 08/16/2022] [Indexed: 11/10/2022]
Abstract
Cholangiocyte death accompanied by the progression of primary biliary cholangitis (PBC) has not yet been thoroughly investigated. Thus, we are aimed to explore the role of HSP90 and a potential treatment strategy in cholangiocyte necroptosis. First, we detected the expression of HSP90 and necroptotic markers in liver tissues from patients and mice with PBC by immunohistochemistry (IHC) and real-time polymerase chain reaction (PCR). Then, the HSP90 inhibitor, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), was administered by intraperitoneal injection to evaluate its therapeutic effect for PBC by IHC, real-time PCR, and western blotting. Human intrahepatic bile duct epithelial cells (HIBECs) were induced to necroptosis by toxic bile acid and lipopolysaccharide (LPS) treatment, and evaluated via Cell Counting Kit-8 and flow cytometry assays. Additionally, 17-DMAG, cycloheximide, and a proteasome inhibitor were used to evaluate the role of HSP90 in cholangiocyte necroptosis. We found that the expression of HSP90 was elevated in the cholangiocytes of patients and mice with PBC, along with higher expressions of receptor-interacting serine/threonine-protein kinase 1 (RIPK1), RIPK3, mixed lineage kinase domain-like protein (MLKL), and phosphorylated-MLKL (p-MLKL). Proinflammatory cytokines and antibody levels of the E2 subunit of pyruvate dehydrogenase complex decreased after treatment with 17-DMAG in PBC mice. Meanwhile, RIPK1, RIPK3, phosphorylated-RIPK3, MLKL, and p-MLKL protein expressions decreased with 17-DMAG treatment. In vitro, 17-DMAG and necrostatin-1 prevented glycochenodeoxycholic acid and LPS-induced necroptosis of HIBECs. Immunoprecipitation and high-performance liquid chromatography-mass spectrometry analysis showed that RIPK1 combined with HSP90. Additionally, the 17-DMAG treatment reduced the RIPK1 half-life. Overall, 17-DMAG might be a potential therapeutic agent for PBC via cholangiocyte necroptosis prevention by accelerating RIPK1 degradation.
Collapse
Affiliation(s)
- Liuying Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiqian Fan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Gastroenterology, General Hospital of the Yangtze River Shipping, Wuhan, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Du
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yixiong Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lilin Hu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhonglin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Mechanisms of Xiaochaihu Decoction on Treating Hepatic Fibrosis Explored by Network Pharmacology. DISEASE MARKERS 2022; 2022:8925637. [PMID: 36246566 PMCID: PMC9553551 DOI: 10.1155/2022/8925637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022]
Abstract
Purpose. To explore the material basis and pharmacological mechanism of Xiaochaihu Decoction (XCHD), the classic Traditional Chinese Medicine (TCM) formula in inhibiting hepatic fibrosis (HF). Methods. The main components in XCHD were screened from the TCMSP database, ETCM database, and literature, and their potential targets were detected and predicted using the Swiss Target Prediction platform. The HF-related targets were retrieved and screened through GeneCard database and OMIM database, combined with GEO gene chips. The XCHD targets and HF targets were mapped to search common targets. The protein-protein interaction (PPI) network was acquired via the STRING11.0 database and analyzed visually using Cytoscape 3.8.0 software. The potential mechanisms of the common targets identified through GO and KEGG pathway enrichment analysis were analyzed by using Metascape database. The results were visualized through OmicShare Tools. The “XCHD compound-HF target” network was visually constructed by Cytoscape 3.8.0 software. AutoDockVina1.1.2 and PyMoL software were used to verify the molecular docking of XCHD main active compounds and HF key targets. Results. A total of 164 potential active compounds from XCHD were screened to act on 95 HF-related targets. Bioinformatics analysis revealed that quercetin, β-sitosterol, and kaempferol may be candidate agents, which acted on multiple targets like PTGS2, HSP90AA1, and PTGS1 and regulate multiple key biological pathways like IL-17 signaling pathway, TNF signaling pathway and PI3K-Akt signaling pathway to relieve HF. Moreover, molecular docking suggested that quercetin and PTGS2 could statically bind and interact with each other through amino acid residues val-349, LEU-352, PHE-381, etc. Conclusion. This work provides a systems perspective to study the relationship between Chinese medicines and diseases. The therapeutic efficacy of XCHD on HF was the sum of multitarget and multi-approach effects from the bioactive ingredients. This study could be one of the cornerstones for further research.
Collapse
|
32
|
Choudhury A, Ratna A, Lim A, Sebastian RM, Moore CL, Filliol AA, Bledsoe J, Dai C, Schwabe RF, Shoulders MD, Mandrekar P. Loss of heat shock factor 1 promotes hepatic stellate cell activation and drives liver fibrosis. Hepatol Commun 2022; 6:2781-2797. [PMID: 35945902 PMCID: PMC9512451 DOI: 10.1002/hep4.2058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 06/24/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022] Open
Abstract
Liver fibrosis is an aberrant wound healing response that results from chronic injury and is mediated by hepatocellular death and activation of hepatic stellate cells (HSCs). While induction of oxidative stress is well established in fibrotic livers, there is limited information on stress-mediated mechanisms of HSC activation. Cellular stress triggers an adaptive defense mechanism via master protein homeostasis regulator, heat shock factor 1 (HSF1), which induces heat shock proteins to respond to proteotoxic stress. Although the importance of HSF1 in restoring cellular homeostasis is well-established, its potential role in liver fibrosis is unknown. Here, we show that HSF1 messenger RNA is induced in human cirrhotic and murine fibrotic livers. Hepatocytes exhibit nuclear HSF1, whereas stellate cells expressing alpha smooth muscle actin do not express nuclear HSF1 in human cirrhosis. Interestingly, despite nuclear HSF1, murine fibrotic livers did not show induction of HSF1 DNA binding activity compared with controls. HSF1-deficient mice exhibit augmented HSC activation and fibrosis despite limited pro-inflammatory cytokine response and display delayed fibrosis resolution. Stellate cell and hepatocyte-specific HSF1 knockout mice exhibit higher induction of profibrogenic response, suggesting an important role for HSF1 in HSC activation and fibrosis. Stable expression of dominant negative HSF1 promotes fibrogenic activation of HSCs. Overactivation of HSF1 decreased phosphorylation of JNK and prevented HSC activation, supporting a protective role for HSF1. Our findings identify an unconventional role for HSF1 in liver fibrosis. Conclusion: Our results show that deficiency of HSF1 is associated with exacerbated HSC activation promoting liver fibrosis, whereas activation of HSF1 prevents profibrogenic HSC activation.
Collapse
Affiliation(s)
- Asmita Choudhury
- Department of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Anuradha Ratna
- Department of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Arlene Lim
- Department of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Rebecca M. Sebastian
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Christopher L. Moore
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Aveline A. Filliol
- Institute of Human NutritionColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Jacob Bledsoe
- Department of PathologyUniversity of Massachusetts Memorial Medical CenterWorcesterMassachusettsUSA
| | - Chengkai Dai
- Center for Cancer Research, National Cancer InstituteFrederickMarylandUSA
| | - Robert F. Schwabe
- Institute of Human NutritionColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Matthew D. Shoulders
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Pranoti Mandrekar
- Department of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| |
Collapse
|
33
|
Zafari N, Velayati M, Fahim M, Maftouh M, Pourali G, Khazaei M, Nassiri M, Hassanian SM, Ghayour-Mobarhan M, Ferns GA, Kiani MA, Avan A. Role of gut bacterial and non-bacterial microbiota in alcohol-associated liver disease: Molecular mechanisms, biomarkers, and therapeutic prospective. Life Sci 2022; 305:120760. [PMID: 35787997 DOI: 10.1016/j.lfs.2022.120760] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 12/17/2022]
Abstract
Alcohol-associated liver disease (ALD) comprises a spectrum of liver diseases that include: steatosis to alcohol-associated hepatitis, cirrhosis, and ultimately hepatocellular carcinoma. The pathophysiology and potential underlying mechanisms for alcohol-associated liver disease are unclear. Moreover, the treatment of ALD remains a challenge. Intestinal microbiota include bacteria, fungi, and viruses, that are now known to be important in the development of ALD. Alcohol consumption can change the gut microbiota and function leading to liver disease. Given the importance of interactions between intestinal microbiota, alcohol, and liver injury, the gut microbiota has emerged as a potential biomarker and therapeutic target. This review focuses on the potential mechanisms by which the gut microbiota may be involved in the pathogenesis of ALD and explains how this can be translated into clinical management. We discuss the potential of utilizing the gut microbiota signature as a biomarker in ALD patients. Additionally, we present an overview of the prospect of modulating the intestinal microbiota for the management of ALD.
Collapse
Affiliation(s)
- Nima Zafari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahla Velayati
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mostafa Fahim
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Mohammad Ali Kiani
- Department of Pediatrics, Akbar Hospital, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pediatric Gastroenterology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Shi X, Li T, Liu Y, Yin L, Xiao L, Fu L, Zhu Y, Chen H, Wang K, Xiao X, Zhang H, Tan S, Tan S. HSF1 Protects Sepsis-Induced Acute Lung Injury by Inhibiting NLRP3 Inflammasome Activation. Front Immunol 2022; 13:781003. [PMID: 35720321 PMCID: PMC9199371 DOI: 10.3389/fimmu.2022.781003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
As an important transcription factor, heat shock factor 1 (HSF1) plays an endogenous anti-inflammation role in the body and can alleviate multiple organ dysfunction caused by sepsis, which contributes to an uncontrolled inflammatory response. The NLRP3 inflammasome is a supramolecular complex that plays key roles in immune surveillance. Inflammation is accomplished by NLRP3 inflammasome activation, which leads to the proteolytic maturation of IL-1β and pyroptosis. However, whether HSF1 is involved in the activation of the NLRP3 inflammasome in septic acute lung injury (ALI) has not been reported. Here, we show that HSF1 suppresses NLRP3 inflammasome activation in transcriptional and post-translational modification levels. HSF1 can repress NLRP3 expression via inhibiting NF-κB phosphorylation. HSF1 can inhibit caspase-1 activation and IL-1β maturation via promoting NLRP3 ubiquitination. Our finding not only elucidates a novel mechanism for HSF1-mediated protection of septic ALI but also identifies new therapeutic targets for septic ALI and related diseases.
Collapse
Affiliation(s)
- Xueyan Shi
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research, Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union, Medical College, Beijing, China
| | - Tao Li
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Medical College of Jiaying University, Meizhou, China
| | - Yanting Liu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Leijin Yin
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lan Xiao
- Department of Traditional Chinese Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Liyao Fu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yaxi Zhu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Huan Chen
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Kangkai Wang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Xianzhong Xiao
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Huali Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Sichuang Tan
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sipin Tan
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, China.,Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
35
|
Liu SX, Liu H, Wang S, Zhang CL, Guo FF, Zeng T. Diallyl disulfide ameliorates ethanol-induced liver steatosis and inflammation by maintaining the fatty acid catabolism and regulating the gut-liver axis. Food Chem Toxicol 2022; 164:113108. [PMID: 35526736 DOI: 10.1016/j.fct.2022.113108] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/12/2022] [Accepted: 04/30/2022] [Indexed: 12/18/2022]
Abstract
Diallyl disulfide (DADS) has been suggested to possess hepatoprotection against alcoholic liver disease (ALD) by a couple of pilot studies, while the underlying mechanisms remain largely unknown. This study aimed to investigate the hepatoprotective effects of DADS against ethanol-induced liver steatosis and early inflammation by using the chronic-plus-binge mice model and cultured J774A.1 macrophages and AML12 hepatocytes. We found that DADS significantly attenuated ethanol-induced elevation of serum aminotransferase activities, accumulation of liver triglyceride, hepatocytes apoptosis, oxidative stress, infiltration of macrophages and neutrophils, and proinflammatory polarization of macrophages in mice livers. In addition, chronic-plus-binge drinking induced apparent intestinal mucosa damage and disturbance of gut microbiota, endotoxemia, and activation of hepatic NF-κB signaling and NLRP3 inflammasome, which was inhibited by DADS. In vitro studies using cocultured AML12/J774A.1 cells showed that DADS suppressed ethanol/LPS-induced cell injury and inflammatory activation of macrophages. Furthermore, DADS ameliorated ethanol-induced decline of peroxisome proliferator-activated receptor α (PPARα), carnitine palmitoyltransferase 1 (CPT1), and phosphorylated AMP-activated protein kinase (AMPK) protein levels in mice liver and AML12 cells. These results demonstrate that DADS could prevent ethanol-induced liver steatosis and early inflammation by regulating the gut-liver axis and maintaining fatty acid catabolism.
Collapse
Affiliation(s)
- Shi-Xuan Liu
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Hong Liu
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shuo Wang
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong Province, 252059, China
| | - Cui-Li Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Fang-Fang Guo
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
36
|
Hong J, Ding J, Hong HH, Xu XW, Pan B, Ruan Y, Zhai XF. Identifying the Mechanism of Polygoni Cuspidati Rhizoma et Radix in Treating Acute Liver Failure Based on Network Pharmacology and Molecular Docking. Gastroenterol Res Pract 2022; 2022:2021066. [PMID: 35432526 PMCID: PMC9012611 DOI: 10.1155/2022/2021066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 12/11/2022] Open
Abstract
Materials and Methods The potential bioactive compounds of PCRR and their targets were collected from TCMSP, TCMID, and BATMAN-TCM databases with absorption, distribution, metabolism, and excretion protocols (oral bioavailability ≥30% and drug-likeness ≥0.18). The ALF-related target genes were identified using the GeneCards and OMIM databases. A protein-protein interaction (PPI) network among these targets was constructed using the Cytoscape software to obtain the core targets. The genes associated with ALF were analyzed via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses to identify the signaling pathways related to the therapeutic effect of PCRR in ALF. Results In total, 10 bioactive compounds of PCRR and 200 targets related to them were obtained, and 2913 ALF-related target genes were identified. PPI network analysis pinpointed 15 core targets, namely, TP53, AKT1, JUN, HSP90AA1, MAPK1, RELA, TNF, ESR1, IL6, MYC, MAPK14, FOS, RB1, CDKN1A, and EGFR. GO enrichment and KEGG pathway analyses revealed that the therapeutic mechanisms of PCRR in ALF are related to cell metabolism, oxidative stress, inflammation, and hepatocyte apoptosis. Conclusion This is the first study to explore the therapeutic mechanisms of PCRR in ALF via network pharmacology and molecular docking. This study provides a research platform with candidate ALF-related targets of PRCC for the development of therapeutics against ALF.
Collapse
Affiliation(s)
- Jing Hong
- Department of Integrative Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Jie Ding
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai 200433, China
- Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Han-han Hong
- Department of Nursing, Chengjiaqiao Community Health Service Center of Changning District, Shanghai 201103, China
| | - Xiao-wan Xu
- Department of Integrative Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Bo Pan
- Department of Integrative Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yi Ruan
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Xiao-feng Zhai
- Department of Integrative Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
37
|
Xia Y, Zhang Q, Ye Y, Wu X, He F, Peng Y, Yin Y, Ren W. Melatonergic signalling instructs transcriptional inhibition of IFNGR2 to lessen interleukin-1β-dependent inflammation. Clin Transl Med 2022; 12:e716. [PMID: 35184395 PMCID: PMC8858632 DOI: 10.1002/ctm2.716] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Immunotransmitters (e.g., neurotransmitters and neuromodulators) could orchestrate diverse immune responses; however, the elaborated mechanism by which melatonergic activation governs inflammation remains less defined. METHODS Primary macrophages, various cell lines, and Pasteurella multocida (PmCQ2)-infected mice were respectively used to illustrate the influence of melatonergic signalling on inflammation in vitro and in vivo. A series of methods (e.g., RNA-seq, metabolomics, and genetic manipulation) were conducted to reveal the mechanism whereby melatonergic signalling reduces macrophage inflammation. RESULTS Here, we demonstrate that melatonergic activation substantially lessens interleukin (IL)-1β-dependent inflammation. Treatment of macrophages with melatonin rewires metabolic program, as well as remodels signalling pathways which depends on interferon regulatory factor (IRF) 7. Mechanistically, melatonin acts via membrane receptor (MT) 1 to increase heat shock factor (Hsf) 1 expression through lowering the inactive glycogen synthase kinase (GSK3) β, thereby transcriptionally inhibiting interferon (IFN)-γ receptor (IFNGR) 2 and ultimately causing defective canonical signalling events [Janus kinase (JAK) 1/2-signal transducer and activator of transcription (STAT) 1-IRF7] and lower IL-1β production in macrophages. Moreover, we find that melatonin amplifies host protective responses to PmCQ2 infection-induced pneumonia. CONCLUSIONS Our conceptual framework provides potential therapeutic targets to prevent and/or treat inflammatory diseases associating with excessive IL-1β production.
Collapse
Affiliation(s)
- Yaoyao Xia
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐BioresourcesGuangdong Laboratory of Lingnan Modern AgricultureGuangdong Provincial Key Laboratory of Animal Nutrition ControlNational Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouChina
| | - Qingzhuo Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐BioresourcesGuangdong Laboratory of Lingnan Modern AgricultureGuangdong Provincial Key Laboratory of Animal Nutrition ControlNational Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouChina
| | - Yuyi Ye
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐BioresourcesGuangdong Laboratory of Lingnan Modern AgricultureGuangdong Provincial Key Laboratory of Animal Nutrition ControlNational Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouChina
| | - Xiaoyan Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐BioresourcesGuangdong Laboratory of Lingnan Modern AgricultureGuangdong Provincial Key Laboratory of Animal Nutrition ControlNational Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouChina
| | - Fang He
- College of Veterinary MedicineSouthwest UniversityChongqingChina
| | - Yuanyi Peng
- College of Veterinary MedicineSouthwest UniversityChongqingChina
| | - Yulong Yin
- Institute of Subtropical AgricultureChinese Academy of SciencesChangshaChina
| | - Wenkai Ren
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐BioresourcesGuangdong Laboratory of Lingnan Modern AgricultureGuangdong Provincial Key Laboratory of Animal Nutrition ControlNational Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouChina
| |
Collapse
|
38
|
Ratna A, Lim A, Li Z, Argemi J, Bataller R, Chiosis G, Mandrekar P. Myeloid Endoplasmic Reticulum Resident Chaperone GP96 Facilitates Inflammation and Steatosis in Alcohol-Associated Liver Disease. Hepatol Commun 2021; 5:1165-1182. [PMID: 34278167 PMCID: PMC8279472 DOI: 10.1002/hep4.1713] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/31/2021] [Accepted: 03/01/2021] [Indexed: 12/17/2022] Open
Abstract
Cellular stress-mediated chaperones are linked to liver macrophage activation and inflammation in alcohol-associated liver disease (ALD). In this study, we investigate the role of endoplasmic reticulum (ER) resident stress chaperone GP96/HSP90B1/GRP94, paralog of the HSP90 family, in ALD pathogenesis. We hypothesize that ER resident chaperone, heat shock protein GP96, plays a crucial role in alcohol-associated liver inflammation and contributes to liver injury. We show high expression of GP96/HSP90B1 and GRP78/HSPA5 in human alcohol-associated hepatitis livers as well as in mouse ALD livers with induction of GP96 prominent in alcohol-exposed macrophages. Myeloid-specific GP96 deficient (M-GP96KO) mice failed to induce alcohol-associated liver injury. Alcohol-fed M-GP96KO mice exhibit significant reduction in steatosis, serum endotoxin, and pro-inflammatory cytokines compared with wild-type mice. Anti-inflammatory cytokines interleukin-10 and transforming growth factor β, as well as activating transcription factor 3 and triggering receptor expressed on myeloid cells 2, markers of restorative macrophages, were higher in alcohol-fed M-GP96KO livers. M-GP96KO mice exhibit protection in a model of endotoxin-mediated liver injury in vivo, which is in agreement with reduced inflammatory responses during ex vivo lipopolysaccharide/endotoxin- stimulated bone marrow-derived macrophages from M-GP96KO mice. Furthermore, we show that liver macrophages from alcohol-fed M-GP96KO mice show compensatory induction of GRP78 messenger RNA, likely due to increased splicing of X-box binding protein-1. Finally, we show that inhibition of GP96 using a specific pharmacological agent, PU-WS13 or small interfering RNA, alleviates inflammatory responses in primary macrophages. Conclusion: Myeloid ER resident GP96 promotes alcohol-induced liver damage through activation of liver macrophage inflammatory responses, alteration in lipid homeostasis, and ER stress. These findings highlight a critical role for liver macrophage ER resident chaperone GP96/HSP90B1 in ALD, and its targeted inhibition represents a promising therapeutic approach in ALD.
Collapse
Affiliation(s)
- Anuradha Ratna
- Department of MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Arlene Lim
- Department of MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Zihai Li
- Division of Medical OncologyDepartment of MedicinePelotonia Institute for Immuno‐OncologyThe Ohio State University Comprehensive Cancer CenterColumbusOHUSA
| | - Josepmaria Argemi
- Division of Gastroenterology, Hepatology and NutritionPittsburgh Liver Research CenterUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Ramon Bataller
- Division of Gastroenterology, Hepatology and NutritionPittsburgh Liver Research CenterUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Gabriela Chiosis
- Chemical Biology ProgramMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Pranoti Mandrekar
- Department of MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| |
Collapse
|
39
|
Le Daré B, Ferron PJ, Gicquel T. The Purinergic P2X7 Receptor-NLRP3 Inflammasome Pathway: A New Target in Alcoholic Liver Disease? Int J Mol Sci 2021; 22:2139. [PMID: 33670021 PMCID: PMC7926651 DOI: 10.3390/ijms22042139] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/24/2022] Open
Abstract
The World Health Organization has estimated that approximately 3 million deaths are attributable to alcohol consumption each year. Alcohol consumption is notably associated with the development and/or progression of many non-communicable inflammatory diseases-particularly in the liver. Although these alcoholic liver diseases were initially thought to be caused by the toxicity of ethanol on hepatocytes, the latest research indicates Kupffer cells (the liver macrophages) are at the heart of this "inflammatory shift". Purinergic signaling (notably through P2X7 receptors and the NLRP3 inflammasome) by Kupffer cells appears to be a decisive factor in the pathophysiology of alcoholic liver disease. Hence, the modulation of purinergic signaling might represent a new means of treating alcoholic liver disease. Here, we review current knowledge on the pathophysiology of alcoholic liver diseases and therapeutic perspectives for targeting these inflammatory pathways.
Collapse
Affiliation(s)
- Brendan Le Daré
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), INSERM, INRAE, CHU—University Rennes, PREVITOX Network, F-35000 Rennes, France; (B.L.D.); (P.-J.F.)
- Forensic and Toxicology Laboratory, Rennes University Hospital, 2 rue Henri Le Guilloux, F-35033 Rennes, France
| | - Pierre-Jean Ferron
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), INSERM, INRAE, CHU—University Rennes, PREVITOX Network, F-35000 Rennes, France; (B.L.D.); (P.-J.F.)
| | - Thomas Gicquel
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), INSERM, INRAE, CHU—University Rennes, PREVITOX Network, F-35000 Rennes, France; (B.L.D.); (P.-J.F.)
- Forensic and Toxicology Laboratory, Rennes University Hospital, 2 rue Henri Le Guilloux, F-35033 Rennes, France
| |
Collapse
|
40
|
Zhang H, Zahid A, Ismail H, Tang Y, Jin T, Tao J. An overview of disease models for NLRP3 inflammasome over-activation. Expert Opin Drug Discov 2020; 16:429-446. [PMID: 33131335 DOI: 10.1080/17460441.2021.1844179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Inflammatory reactions, including those mediated by the NLRP3 inflammasome, maintain the body's homeostasis by removing pathogens, repairing damaged tissues, and adapting to stressed environments. However, uncontrolled activation of the NLRP3 inflammasome tends to cause various diseases using different mechanisms. Recently, many inhibitors of the NLRP3 inflammasome have been reported and many are being developed. In order to assess their efficacy, specificity, and mechanism of action, the screening process of inhibitors requires various types of cell and animal models of NLRP3-associated diseases.Areas covered: In the following review, the authors give an overview of the cell and animal models that have been used during the research and development of various inhibitors of the NLRP3 inflammasome.Expert opinion: There are many NLRP3 inflammasome inhibitors, but most of the inhibitors have poor specificity and often influence other inflammatory pathways. The potential risk for cross-reaction is high; therefore, the development of highly specific inhibitors is essential. The selection of appropriate cell and animal models, and combined use of different models for the evaluation of these inhibitors can help to clarify the target specificity and therapeutic effects, which is beneficial for the development and application of drugs targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ayesha Zahid
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hazrat Ismail
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science. Hefei National Science Center for Physical Sciences at Microscale. University of Science and Technology of China, Hefei, China
| | - Yujie Tang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Shanghai, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
41
|
Zhang F, Zhao W, Zhou J, Wang W, Luo J, Feng Y, Wu J, Li M, Wang K, Niu J, Miao Y. Heat shock transcription factor 2 reduces the secretion of IL-1β by inhibiting NLRP3 inflammasome activation in ulcerative colitis. Gene 2020; 768:145299. [PMID: 33181254 DOI: 10.1016/j.gene.2020.145299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/25/2020] [Accepted: 11/04/2020] [Indexed: 10/23/2022]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease with unknown aetiology. As a pro-inflammatory cytokine, interleukin-1β (IL-1β) plays a critical, damaging role in UC. Heat shock proteins (HSPs) are important anti-inflammatory factors that maintain intestinal epithelial cells (IECs) homeostasis. Heat shock transcription factor 2 (HSF2) is an important regulator of HSPs. In our previous research, we found that HSF2 is highly expressed in UC, is negatively related to colon inflammation of mice, and inhibits the expression of IL-1β, but the specific mechanism is still unclear. As a product of the NLRP3 inflammasome, the expression of IL-1β is closely related to NLRP3 inflammasome activation. Therefore, we hypothesised that HSF2 affects the secretion of IL-1β by regulating activation of the NLRP3 inflammasome. In this study, hsf-/- DSS model mice showed highest levels of expression of the NLRP3 inflammasome and the secretion of IL-1β. In Caco-2 cells, the levels of expression of the NLRP3 inflammasome and the secretion of IL-1β were inhibited by overexpression of HSF2, and inhibited HSF2 increased activation of the NLRP3 inflammasome and the secretion of IL-1β. These findings indicated that HSF2 might be an important target for inflammatory modulation in UC.
Collapse
Affiliation(s)
- Fengrui Zhang
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming 650032, China
| | - Wei Zhao
- Kunming Medical University, Kunming 650032, China
| | - Jiao Zhou
- Kunming Medical University, Kunming 650032, China
| | - Wen Wang
- Kunming Medical University, Kunming 650032, China
| | - Juan Luo
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming 650032, China
| | - Yuran Feng
- Department of Ultrasound, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Jing Wu
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming 650032, China
| | - Maojuan Li
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming 650032, China
| | - Kunhua Wang
- Department of General Surgery, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming 650032, China
| | - Junkun Niu
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming 650032, China.
| | - Yinglei Miao
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming 650032, China.
| |
Collapse
|
42
|
Nizami S, Arunasalam K, Green J, Cook J, Lawrence CB, Zarganes-Tzitzikas T, Davis JB, Di Daniel E, Brough D. Inhibition of the NLRP3 inflammasome by HSP90 inhibitors. Immunology 2020; 162:84-91. [PMID: 32954500 PMCID: PMC7730016 DOI: 10.1111/imm.13267] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/25/2020] [Accepted: 09/09/2020] [Indexed: 12/19/2022] Open
Abstract
Excessive and dysregulated inflammation is known to contribute to disease progression. HSP90 is an intracellular chaperone known to regulate inflammatory processes including the NLRP3 inflammasome and secretion of the pro‐inflammatory cytokine interleukin(IL)‐1β. Here, primarily using an in vitro inflammasome ASC speck assay, and an in vivo model of murine peritonitis, we tested the utility of HSP90 inhibitors as anti‐inflammatory molecules. We report that the HSP90 inhibitor EC144 effectively inhibited inflammatory processes including priming and activation of NLRP3 in vitro and in vivo. A specific inhibitor of the β HSP90 isoform was ineffective suggesting the importance of the α isoform in inflammatory signalling. EC144 inhibited IL‐1β and IL‐6 in vivo when administered orally, and was brain‐penetrant. These data suggest that HSP90 inhibitors may be useful for targeting inflammation in diverse diseases that are worsened by the presence of inflammation.
Collapse
Affiliation(s)
- Sohaib Nizami
- Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, UK
| | - Kanisa Arunasalam
- Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, UK
| | - Jack Green
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - James Cook
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Catherine B Lawrence
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | | | - John B Davis
- Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, UK
| | - Elena Di Daniel
- Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, UK
| | - David Brough
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| |
Collapse
|
43
|
Wang X, Fan L, Yin H, Zhou Y, Tang X, Fei X, Tang H, Peng J, Ren X, Xue Y, Zhu C, Luo J, Jin Q, Jin Q. Protective effect of Aster tataricus extract on NLRP3-mediated pyroptosis of bladder urothelial cells. J Cell Mol Med 2020; 24:13336-13345. [PMID: 33030301 PMCID: PMC7701514 DOI: 10.1111/jcmm.15952] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/17/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022] Open
Abstract
Aster tataricus L.f. is a traditional Eastern Asian herbal medicine used for the relief of uroschesis‐related illnesses and has been demonstrated clinically to exert satisfied effects. However, the mechanism of its therapeutic action remains unclear. The present study aimed to evaluate the protective mechanism of Aster tataricus extract (ATE) on CYP or LPS + ATP‐induced interstitial cystitis (IC), we successfully constructed the induced IC Sprague‐Dawley (SD) rat model and IC human urothelium cell (SV‐HUC‐1) model. The main compounds of ATE were determined by LC‐MS. After intervention, the changes on the bladder wall morphology and inflammation were observed in each group. SV‐HUC1 cell viability was measured by MTT and double stained with Hoechst 33342 and propidium iodide (PI). The expression levels of NLRP3, Pro‐caspase‐1, Caspsae‐1 p20, GSDMD, GSDMD‐N and Cleave‐IL‐1β in vivo and in vitro in different groups were detected by Western blotting. ATE significantly alleviated oedema and haemorrhage and reduced the inflammation index and histopathological score in SD rat bladder. The results of cell revealed that ATE could improve cell viability and decrease pyroptosis ratio. The expression of NLRP3 and other pyroptosis‐related protein was remarkably decreased by ATE both in vivo and in vitro. ATE may be used as an inhibitor of NLRP3 in treating IC. The discovery of NLRP3/Caspase‐1/GSDMD‐N as a new protective pathway provides a new direction for protecting cell against IC.
Collapse
Affiliation(s)
- Xin Wang
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China.,Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Ling Fan
- Department of Pharmacy, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Hao Yin
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China.,Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Yiqun Zhou
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Xiaolong Tang
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Xiaojun Fei
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Hailin Tang
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Juan Peng
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Xiaoqin Ren
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Yi Xue
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Chunli Zhu
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Jianping Luo
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Qinglei Jin
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Qingjiang Jin
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| |
Collapse
|
44
|
Molecular Chaperones: Molecular Assembly Line Brings Metabolism and Immunity in Shape. Metabolites 2020; 10:metabo10100394. [PMID: 33023034 PMCID: PMC7600384 DOI: 10.3390/metabo10100394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Molecular chaperones are a set of conserved proteins that have evolved to assist the folding of many newly synthesized proteins by preventing their misfolding under conditions such as elevated temperatures, hypoxia, acidosis and nutrient deprivation. Molecular chaperones belong to the heat shock protein (HSP) family. They have been identified as important participants in immune functions including antigen presentation, immunostimulation and immunomodulation, and play crucial roles in metabolic rewiring and epigenetic circuits. Growing evidence has accumulated to indicate that metabolic pathways and their metabolites influence the function of immune cells and can alter transcriptional activity through epigenetic modification of (de)methylation and (de)acetylation. However, whether molecular chaperones can regulate metabolic programs to influence immune activity is still largely unclear. In this review, we discuss the available data on the biological function of molecular chaperones to immune responses during inflammation, with a specific focus on the interplay between molecular chaperones and metabolic pathways that drive immune cell fate and function.
Collapse
|
45
|
Harnessing the Proteostasis Network in Alcohol-associated Liver Disease. CURRENT PATHOBIOLOGY REPORTS 2020. [DOI: 10.1007/s40139-020-00211-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
46
|
Yao L, Song J, Meng XW, Ge JY, Du BX, Yu J, Ji FH. Periostin aggravates NLRP3 inflammasome-mediated pyroptosis in myocardial ischemia-reperfusion injury. Mol Cell Probes 2020; 53:101596. [PMID: 32461137 DOI: 10.1016/j.mcp.2020.101596] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
Pyroptosis is a form of caspase-1-induced programmed cell death. This study aimed to investigate the effect of periostin (postn) on pyroptosis in myocardial ischemia-reperfusion injury (MIRI). To this end, the differentially expressed genes were obtained from the GSE4105 dataset using the "GEO2R" online tool. Protein-protein interaction networks were constructed using the Search Tool for the Retrieval of Interacting Genes (STRING) database, and Module and Go analysis were conducted using the Cytoscape 3.6 plugs-in MCODE and BINGO, respectively. The analysis showed that postn was a critical gene in the most significant module. Experimental results, including triphenyltetrazolium chloride staining, pathological analysis, TUNEL staining, western blotting, and RT-qPCR assays, showed that MIRI induced caspase-1-mediated pyroptosis by activating the NLRP3 inflammasome. Postn was significantly upregulated in the heart tissues of MIRI rats and in H9C2 cells following hypoxia/reoxygenation (H/R) treatment. In addition, knockdown of postn suppressed the caspase-1-mediated pyroptosis and H/R-mediated NLRP3 inflammasome activation, as evidenced by flow cytometry, CCK8, RT-qPCR, western blotting, and ELISA assays. In contrast, overexpression of postn promoted NLRP3 inflammasome-mediated pyroptosis of H/R-treated H9C2 cells. According to the results of rescue experiments, a caspase-1 inhibitor reduced the increase in NLRP3 inflammasome-mediated pyroptosis induced by overexpression of postn, and the pyroptosis-promoting function of postn overexpression in H/R treated H9C2 cells was reversed by inhibition of NLRP3. In conclusion, postn overexpression promoted the caspase-1-mediated pyroptosis during MIRI by activating the NLRP3.
Collapse
Affiliation(s)
- Lei Yao
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, PR China; Department of Anesthesiology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Jie Song
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Xiao Wen Meng
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, PR China
| | - Jian Yun Ge
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Bo Xiang Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Jun Yu
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, PR China
| | - Fu Hai Ji
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, PR China.
| |
Collapse
|