1
|
Chen Y, Liu J, Sun Y, Li M, Fan X, Gu X. Multi-omics study reveals Shuangshen Pingfei formula regulates EETs metabolic reprogramming to exert its therapeutic effect on pulmonary fibrosis. Int Immunopharmacol 2024; 143:113275. [PMID: 39395378 DOI: 10.1016/j.intimp.2024.113275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/05/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024]
Abstract
As a clinical formula derived from Renshen Pingfei San, Shuangshen Pingfei formula (SSPF) has been used to treat pulmonary fibrosis (PF). However, its in-depth mechanism of action remains unknown. In this study, the effect of SSPF was evaluated by applying a rat model of PF caused by intratracheal drip bleomycin. To characterize the molecular changes related to PF and reveal therapeutic targets for SSPF, we performed transcriptomic and metabolomic analyses on rat lung. Finally, western blotting and qPCR experiments were used to validate the multi-omics results. As a result, a significant reduction in inflammation and fibrosis caused by BLM was observed when SSPF was administered. Widespread changes in gene expression and metabolic programming were observed in the lungs of PF rats through RNA-seq and untargeted metabolomic analysis. Combined transcriptomic and metabolomic analyses revealed the involvement of arachidonic acid (AA) metabolism pathways in PF. Further validation of AA metabolite synthase genes and protein levels showed a significant decrease in the levels of epoxyeicosatrienoic acids (EETs) synthases, including Cyp2j2, Cyp2b1, in the PF lungs. SSPF treatment regulated the above changes in gene expression and metabolic programming, particularly the regulation of EETs. This study is the first to investigate the mechanism of action of SSPF in the treatment of PF from the perspective of regulating the synthesis of EETs in AA metabolism.
Collapse
Affiliation(s)
- Yeqing Chen
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jiayi Liu
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yubo Sun
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengwen Li
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinsheng Fan
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xin Gu
- College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Weng J, Cheng Q, Yang J, Jin H, Zhang R, Guan J, Ma Y, Wang L, Chen C, Wang Z. Gal-1-mediated cytochrome p450 activation promotes fibroblast into myofibroblast differentiation in pulmonary fibrosis. Int Immunopharmacol 2024; 141:112920. [PMID: 39137631 DOI: 10.1016/j.intimp.2024.112920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Pulmonary fibrosis (PF) results from excessive extracellular matrix (ECM) deposition and tissue remodeling after activation of fibroblasts into myofibroblasts. Abnormally deposited fibrotic ECM, in turn, promotes fibroblast activation and accelerates loss of lung structure and function. However, the molecular mediators and exact mechanisms by which fibrotic ECM promotes fibroblast activation are unclear. In a bleomycin-induced PF mouse model, we found Galectin-1 (Gal-1) expression was significantly increased in lung tissue, and overexpression of Gal-1 plasmid-transfected fibroblasts were activated into myofibroblasts. Using the decellularization technique to prepare decellularized fibrotic ECM and constructing a 3D in vitro co-culture system with fibroblasts, we found that decellularized fibrotic ECM induced a high expression of Gal-1 and promoted the activation of fibroblasts into myofibroblasts. Therefore, Gal-1 has been identified as a pivotal mediator in PF. Further, we found that decellularized fibrotic ECM delivered mechanical signals to cells through the Gal-1-mediated FAK-Src-P130Cas mechanical signalling pathway, while the CYP450 enzymes (mainly involved in CYP1A1, CYP24A1, CYP3A4, and CYP2D6 isoforms) acted as a chemical signalling pathway to receive mechanical signals transmitted from upstream Gal-1, thereby promoting fibroblast activation. The Gal-1 inhibitor OTX008 or the CYP1A1 inhibitor 7-Hydroxyflavone prevented PF in mice and inhibited the role of fibrotic ECM in promoting fibroblast activation into myofibroblasts, preventing PF. These results reveal novel molecular mechanisms of lung fibrosis formation and identify Gal-1 and its downstream CYP1A1 as potential therapeutic targets for PF disease treatmnts.
Collapse
Affiliation(s)
- Jie Weng
- Department of General Practice, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou 325000, China; South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou 325014, China
| | - Qianhui Cheng
- Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jingwen Yang
- Department of General Practice, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou 325000, China; Department of General Practice, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou 318001, China
| | - Haijuan Jin
- Department of General Practice, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Theorem Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, China
| | - Ran Zhang
- Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiangan Guan
- Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuan Ma
- Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Liang Wang
- Department of Public Health, Robbins College of Health and Human Sciences, Baylor University, Waco, TX, USA
| | - Chan Chen
- Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou 325000, China; Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Zhiyi Wang
- Department of General Practice, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou 325000, China; South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou 325014, China; Department of General Practice, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou 318001, China.
| |
Collapse
|
3
|
Wu H, Li D, Zhang CY, Huang LL, Zeng YJ, Chen TG, Yu K, Meng JW, Lin YX, Guo R, Zhou Y, Gao G. Restoration of ARA metabolic disorders in vascular smooth muscle cells alleviates intimal hyperplasia. Eur J Pharmacol 2024; 983:176824. [PMID: 39265882 DOI: 10.1016/j.ejphar.2024.176824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/14/2024]
Abstract
Intimal hyperplasia (IH) is an innegligible issue for patients undergoing interventional therapy. The proliferation and migration of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor-BB (PDGF-BB) are critical events in the development of IH. While the exact mechanism and effective target for IH needs further investigation. Metabolic disorders of arachidonic acid (ARA) are involved in the occurrence and progression of various diseases. In this study, we found that the expressions of soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2) were significantly increased in the VSMCs during balloon injury-induced IH. Then, we employed a COX-2/sEH dual inhibitor PTUPB to increase the concentration of epoxyeicosatrienoic acids (EETs) while prevent the release of pro-inflammatory prostaglandins. Results showed that PTUPB treatment significantly reduced neointimal thickening induced by balloon injury in rats in vivo and inhibited PDGF-BB-induced proliferation and migration of VSMCs in vitro. Our results showed that PTUPB may reverse the phenotypic transition of VSMCs by inhibiting Pttg1 expression. In conclusion, we found that the dysfunction of ARA metabolism in VSMCs contributes to IH, and the COX-2/sEH dual inhibitor PTUPB attenuates IH progression by reversing the phenotypic switch in VSMC through the Sirt1/Pttg1 pathway.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Hyperplasia
- Male
- Rats
- Cyclooxygenase 2/metabolism
- Cell Proliferation/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Rats, Sprague-Dawley
- Cell Movement/drug effects
- Epoxide Hydrolases/antagonists & inhibitors
- Epoxide Hydrolases/metabolism
- Tunica Intima/pathology
- Tunica Intima/metabolism
- Tunica Intima/drug effects
- Becaplermin/pharmacology
- Neointima/pathology
- Neointima/metabolism
- Neointima/drug therapy
- Metabolic Diseases/metabolism
- Metabolic Diseases/drug therapy
- Metabolic Diseases/pathology
Collapse
Affiliation(s)
- Hui Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Dai Li
- Phase I Clinical Research Center, Xiangya Hospital, Central South University, Changsha, 410005, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Ling-Li Huang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - You-Jie Zeng
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Tian-Ge Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Ke Yu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jia-Wei Meng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yu-Xin Lin
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Ren Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, China.
| | - Ge Gao
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
4
|
Pitchai A, Shinde A, Swihart JN, Robison K, Shannahan JH. Specialized Pro-Resolving Lipid Mediators Distinctly Modulate Silver Nanoparticle-Induced Pulmonary Inflammation in Healthy and Metabolic Syndrome Mouse Models. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1642. [PMID: 39452978 PMCID: PMC11510677 DOI: 10.3390/nano14201642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024]
Abstract
Individuals with chronic diseases are more vulnerable to environmental inhalation exposures. Although metabolic syndrome (MetS) is increasingly common and is associated with susceptibility to inhalation exposures such as particulate air pollution, the underlying mechanisms remain unclear. In previous studies, we determined that, compared to a healthy mouse model, a mouse model of MetS exhibited increased pulmonary inflammation 24 h after exposure to AgNPs. This exacerbated response was associated with decreases in pulmonary levels of specific specialized pro-resolving mediators (SPMs). Supplementation with specific SPMs that are known to be dysregulated in MetS may alter particulate-induced inflammatory responses and be useful in treatment strategies. Our current study hypothesized that administration of resolvin E1 (RvE1), protectin D1 (PD1), or maresin (MaR1) following AgNP exposure will differentially regulate inflammatory responses. To examine this hypothesis, healthy and MetS mouse models were exposed to either a vehicle (control) or 50 μg of 20 nm AgNPs via oropharyngeal aspiration. They were then treated 24 h post-exposure with either a vehicle (control) or 400 ng of RvE1, PD1, or MaR1 via oropharyngeal aspiration. Endpoints of pulmonary inflammation and toxicity were evaluated three days following AgNP exposure. MetS mice that were exposed to AgNPs and received PBS treatment exhibited significantly exacerbated pulmonary inflammatory responses compared to healthy mice. In mice exposed to AgNPs and treated with RvE1, neutrophil infiltration was reduced in healthy mice and the exacerbated neutrophil levels were decreased in the MetS model. This decreased neutrophilia was associated with decreases in proinflammatory cytokines' gene and protein expression. Healthy mice treated with PD1 did not demonstrate alterations in AgNP-induced neutrophil levels compared to mice not receiving treat; however, exacerbated neutrophilia was reduced in the MetS model. These PD1 alterations were associated with decreases in proinflammatory cytokines, as well as elevated interleukin-10 (IL-10). Both mouse models receiving MaR1 treatment demonstrated reductions in AgNP-induced neutrophil influx. MaR1 treatment was associated with decreases in proinflammatory cytokines in both models and increases in the resolution inflammatory cytokine IL-10 in both models, which were enhanced in MetS mice. Inflammatory responses to particulate exposure may be treated using specific SPMs, some of which may benefit susceptible subpopulations.
Collapse
Affiliation(s)
| | | | | | | | - Jonathan H. Shannahan
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
5
|
Jin L, Bao B, Huang XT, Tao JH, Duan JX, Zhong WJ, Zhang CY, Liu YB, Chen H, Yang NSY, Guan CX, Zhou Y. MEOX1 triggers myofibroblast apoptosis resistance, contributing to pulmonary fibrosis in mice. J Cell Physiol 2024:e31442. [PMID: 39319990 DOI: 10.1002/jcp.31442] [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: 06/06/2024] [Revised: 08/22/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
The apoptosis resistance of myofibroblasts is a hallmark in the irreversible progression of pulmonary fibrosis (PF). While the underlying molecular mechanism remains elusive. In this study, we unveiled a previously unrecognized mechanism underlying myofibroblast apoptosis resistance during PF. Our investigation revealed heightened expression of mesenchyme homeobox 1 (MEOX1) in the lungs of idiopathic pulmonary fibrosis (IPF) patients and bleomycin-induced PF mice. Silencing MEOX1 significantly attenuated PF progression in mice. In vitro, we found a notable increase in MEOX1 expression in transforming growth factor-β1 (TGF-β1)-induced myofibroblasts. Silencing MEOX1 enhanced apoptosis of myofibroblasts. Mechanistically, we identified G-protein signaling pathway regulatory factor 4 (RGS4) as a critical downstream target of MEOX1, as predicted by bioinformatics analysis. MEOX1 enhanced apoptosis resistance by upregulating RGS4 expression in myofibroblasts. In conclusion, our study highlights MEOX1 as a promising therapeutic target for protecting against PF by modulating myofibroblast apoptosis resistance.
Collapse
Affiliation(s)
- Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Bo Bao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, China
| | - Jia-Hao Tao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Jia-Xi Duan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Wen-Jin Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Hui Chen
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| |
Collapse
|
6
|
Zhong WJ, Zhang CY, Duan JX, Chen MR, Ping-Deng, Zhang BL, Yang NSY, Sha HX, Zhang J, Xiong JB, Guan CX, Zhou Y. Krüppel-like transcription factor 14 alleviates alveolar epithelial cell senescence by inhibiting endoplasmic reticulum stress in pulmonaryfibrosis. Int J Biol Macromol 2024; 280:135351. [PMID: 39270890 DOI: 10.1016/j.ijbiomac.2024.135351] [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: 07/10/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Pulmonary fibrosis (PF) is defined as a specific form of chronic, progressive fibrosing interstitial pneumonia, occurring primarily in older adults with poor prognosis. Alveolar epithelial cell (AEC) senescence is the critical pathological mechanism of PF. However, the molecular mechanisms regulating AEC senescence in PF are incompletely understood. Herein, we provided evidence to support the function of Krüppel-like factor 14 (KLF14), a novel Krüppel-like transcription factor, in the regulation of AEC senescence during PF. We confirmed that the expression of KLF14 was up-regulated in PF patients and mice treated with bleomycin (BLM). KLF14 knockdown resulted in more pronounced structural disruption of the lung tissue and swelling of the alveolar septum, which led to significantly increased mortality in BLM-induced PF mice. Mechanistically, RNA-seq analysis indicated that KLF14 decreased the senescence of AECs by inhibiting endoplasmic reticulum (ER) stress. Furthermore, the pharmacological activation of KLF14 conferred protection against PF in mice. In conclusion, our findings reveal a protective role for KLF14 in preventing AECs from senescence and shed light on the development of KLF14-targeted therapeutics for PF.
Collapse
Affiliation(s)
- Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Meng-Rui Chen
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Ping-Deng
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Bo-Liang Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Han-Xi Sha
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Jun Zhang
- Department of Physiology, Hunan University of Medicine, Huaihua, China
| | - Jian-Bing Xiong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China.
| |
Collapse
|
7
|
Zeng Q, Gong Y, Zhu N, Shi Y, Zhang C, Qin L. Lipids and lipid metabolism in cellular senescence: Emerging targets for age-related diseases. Ageing Res Rev 2024; 97:102294. [PMID: 38583577 DOI: 10.1016/j.arr.2024.102294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/09/2024]
Abstract
Cellular senescence is a kind of cellular state triggered by endogenous or exogenous stimuli, which is mainly characterized by stable cell cycle arrest and complex senescence-associated secretory phenotype (SASP). Once senescent cells accumulate in tissues, they may eventually accelerate the progression of age-related diseases, such as atherosclerosis, osteoarthritis, chronic lung diseases, cancers, etc. Recent studies have shown that the disorders of lipid metabolism are not only related to age-related diseases, but also regulate the cellular senescence process. Based on existing research evidences, the changes in lipid metabolism in senescent cells are mainly concentrated in the metabolic processes of phospholipids, fatty acids and cholesterol. Obviously, the changes in lipid-metabolizing enzymes and proteins involved in these pathways play a critical role in senescence. However, the link between cellular senescence, changes in lipid metabolism and age-related disease remains to be elucidated. Herein, we summarize the lipid metabolism changes in senescent cells, especially the senescent cells that promote age-related diseases, as well as focusing on the role of lipid-related enzymes or proteins in senescence. Finally, we explore the prospect of lipids in cellular senescence and their potential as drug targets for preventing and delaying age-related diseases.
Collapse
Affiliation(s)
- Qing Zeng
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Yongzhen Gong
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Neng Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410021, China
| | - Yaning Shi
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Chanjuan Zhang
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Institutional Key Laboratory of Vascular Biology and Translational Medicine in Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
8
|
Yousef A, Sosnowski DK, Fang L, Legaspi RJ, Korodimas J, Lee A, Magor KE, Seubert JM. Cardioprotective response and senescence in aged sEH null female mice exposed to LPS. Am J Physiol Heart Circ Physiol 2024; 326:H1366-H1385. [PMID: 38578240 DOI: 10.1152/ajpheart.00706.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Deterioration of physiological systems, like the cardiovascular system, occurs progressively with age impacting an individual's health and increasing susceptibility to injury and disease. Cellular senescence has an underlying role in age-related alterations and can be triggered by natural aging or prematurely by stressors such as the bacterial toxin lipopolysaccharide (LPS). The metabolism of polyunsaturated fatty acids by CYP450 enzymes produces numerous bioactive lipid mediators that can be further metabolized by soluble epoxide hydrolase (sEH) into diol metabolites, often with reduced biological effects. In our study, we observed age-related cardiac differences in female mice, where young mice demonstrated resistance to LPS injury, and genetic deletion or pharmacological inhibition of sEH using trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid attenuated LPS-induced cardiac dysfunction in aged female mice. Bulk RNA-sequencing analyses revealed transcriptomics differences in aged female hearts. The confirmatory analysis demonstrated changes to inflammatory and senescence gene markers such as Il-6, Mcp1, Il-1β, Nlrp3, p21, p16, SA-β-gal, and Gdf15 were attenuated in the hearts of aged female mice where sEH was deleted or inhibited. Collectively, these findings highlight the role of sEH in modulating the aging process of the heart, whereby targeting sEH is cardioprotective.NEW & NOTEWORTHY Soluble epoxide hydrolase (sEH) is an essential enzyme for converting epoxy fatty acids to their less bioactive diols. Our study suggests deletion or inhibition of sEH impacts the aging process in the hearts of female mice resulting in cardioprotection. Data indicate targeting sEH limits inflammation, preserves mitochondria, and alters cellular senescence in the aged female heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Liye Fang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Renald James Legaspi
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jacob Korodimas
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andy Lee
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Katharine E Magor
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Li Q, Sun J, Ran Q, Liu Z, Chen J. The protective effects of Chromofungin in oligomeric amyloid β 42 (Aβ 42)-induced toxicity in neurons in Alzheimer's disease. Aging (Albany NY) 2024; 16:9216-9227. [PMID: 38795392 PMCID: PMC11164494 DOI: 10.18632/aging.205865] [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: 12/30/2023] [Accepted: 04/16/2024] [Indexed: 05/27/2024]
Abstract
Oligomeric Aβ42 is considered to play a harmful role in the pathophysiology of Alzheimer's disease (AD). Prolonged exposure to oligomeric Aβ42 could induce neuronal damage including cellular senescence. Amelioration of Aβ42-induced cellular senescence has been considered as a promising strategy for the treatment of AD. Chromofungin, a chromogranin A-derived peptide, has displayed various biological functions in different types of cells and tissues. However, the effects of Chromofungin on oligomeric Aβ42-induced cellular senescence have not been previously reported. In the current study, we report a novel function of Chromofungin by showing that treatment with Chromofungin could ameliorate Aβ42-induced neurotoxicity in M17 neuronal cells. The Cell Counting Kit-8 (CCK-8) assay and the lactate dehydrogenase (LDH) release experiments revealed that 0.5 and 1 mM are the optimal concentrations of Chromofungin for cell culture in M17 cells. Challenging with oligomeric Aβ42 (5 μM) for 7 and 14 days led to a significant decrease in telomerase activity, which was rescued by Chromofungin dose-dependently. Additionally, the senescence-associated β-galactosidase (SA-β-gal) staining assay demonstrated that Chromofungin mitigated oligomeric Aβ42-induced cellular senescence. Correspondingly, treatment with Chromofungin reversed the gene expression of human telomerase reverse transcriptase (hTERT), telomeric repeat-binding factor 2 (TERF2), and p21 against oligomeric Aβ42 in M17 neurons. Interestingly, Chromofungin attenuated oligomeric Aβ42-induced oxidative stress (OS) in M17 cells by reducing the production of intracellular reactive oxygen species (ROS) but increasing the levels of intracellular superoxide dismutase (SOD). Importantly, the presence of Chromofungin reduced the expression of cyclooxygenase2 (COX-2) as well as the generation of prostaglandin E2 (PGE2). Transduction with Ad-COX-2 impaired the effects of Chromofungin on telomerase activity and the profile of cellular senescence. Our findings suggest that Chromofungin might act as a potential agent for the treatment of AD.
Collapse
Affiliation(s)
- Qingwei Li
- Department of Psychiatry, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ji Sun
- Department of Neurology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China
| | - Qin Ran
- Department of Psychiatry, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ziming Liu
- Department of Psychiatry, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jinmei Chen
- Department of Neurology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China
| |
Collapse
|
10
|
Qian C, Wang Q, Qiao Y, Xu Z, Zhang L, Xiao H, Lin Z, Wu M, Xia W, Yang H, Bai J, Geng D. Arachidonic acid in aging: New roles for old players. J Adv Res 2024:S2090-1232(24)00180-2. [PMID: 38710468 DOI: 10.1016/j.jare.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Arachidonic acid (AA), one of the most ubiquitous polyunsaturated fatty acids (PUFAs), provides fluidity to mammalian cell membranes. It is derived from linoleic acid (LA) and can be transformed into various bioactive metabolites, including prostaglandins (PGs), thromboxanes (TXs), lipoxins (LXs), hydroxy-eicosatetraenoic acids (HETEs), leukotrienes (LTs), and epoxyeicosatrienoic acids (EETs), by different pathways. All these processes are involved in AA metabolism. Currently, in the context of an increasingly visible aging world population, several scholars have revealed the essential role of AA metabolism in osteoporosis, chronic obstructive pulmonary disease, and many other aging diseases. AIM OF REVIEW Although there are some reviews describing the role of AA in some specific diseases, there seems to be no or little information on the role of AA metabolism in aging tissues or organs. This review scrutinizes and highlights the role of AA metabolism in aging and provides a new idea for strategies for treating aging-related diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW As a member of lipid metabolism, AA metabolism regulates the important lipids that interfere with the aging in several ways. We present a comprehensivereviewofthe role ofAA metabolism in aging, with the aim of relieving the extreme suffering of families and the heavy economic burden on society caused by age-related diseases. We also collected and summarized data on anti-aging therapies associated with AA metabolism, with the expectation of identifying a novel and efficient way to protect against aging.
Collapse
Affiliation(s)
- Chen Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Qing Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Ze Xu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China
| | - Linlin Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China
| | - Haixiang Xiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Zhixiang Lin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Mingzhou Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 17 Lujiang Road, Hefei, Anhui 230031, PR China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
11
|
Duan JX, Guan XX, Cheng W, Deng DD, Chen P, Liu C, Zhou Y, Hammock BD, Yang HH. COX-2/sEH-Mediated Macrophage Activation Is a Target for Pulmonary Protection in Mouse Models of Chronic Obstructive Pulmonary Disease. J Transl Med 2024; 104:100319. [PMID: 38158123 DOI: 10.1016/j.labinv.2023.100319] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024] Open
Abstract
Effective inhibition of macrophage activation is critical for resolving inflammation and restoring pulmonary function in patients with chronic obstructive pulmonary disease (COPD). In this study, we identified the dual-enhanced cyclooxygenase-2 (COX-2)/soluble epoxide hydrolase (sEH) as a novel regulator of macrophage activation in COPD. Both COX-2 and sEH were found to be increased in patients and mice with COPD and in macrophages exposed to cigarette smoke extract. Pharmacological reduction of the COX-2 and sEH by 4-(5-phenyl-3-{3-[3-(4-trifluoromethylphenyl)-ureido]-propyl}-pyrazol-1-yl)-benzenesulfonamide (PTUPB) effectively prevented macrophage activation, downregulated inflammation-related genes, and reduced lung injury, thereby improving respiratory function in a mouse model of COPD induced by cigarette smoke and lipopolysaccharide. Mechanistically, enhanced COX-2/sEH triggered the activation of the NACHT, LRR, and PYD domains-containing protein 3 inflammasome, leading to the cleavage of pro-IL-1β into its active form in macrophages and amplifying inflammatory responses. These findings demonstrate that targeting COX-2/sEH-mediated macrophage activation may be a promising therapeutic strategy for COPD. Importantly, our data support the potential use of the dual COX-2 and sEH inhibitor PTUPB as a therapeutic drug for the treatment of COPD.
Collapse
Affiliation(s)
- Jia-Xi Duan
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xin-Xin Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Wei Cheng
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Ding-Ding Deng
- Department of Respiratory Medicine, First Affiliated People's Hospital of Shaoyang College, Shaoyang, China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Cong Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, California
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China.
| |
Collapse
|
12
|
Hong JR, Zhang CY, Zhong WJ, Yang HH, Xiong JB, Deng P, Yang NSY, Chen H, Jin L, Guan CX, Duan JX, Zhou Y. Epoxyeicosatrienoic acids alleviate alveolar epithelial cell senescence by inhibiting mitophagy through NOX4/Nrf2 pathway. Biomed Pharmacother 2023; 169:115937. [PMID: 38007934 DOI: 10.1016/j.biopha.2023.115937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023] Open
Abstract
Alveolar epithelial cell (AEC) senescence is considered to be a universal pathological feature of many chronic pulmonary diseases. Our previous study found that epoxyeicosatrienoic acids (EETs), produced from arachidonic acid (ARA) through the cytochrome P450 cyclooxygenase (CYP) pathway, have significant negative regulatory effects on cellular senescence in AECs. However, the exact mechanisms by which EETs alleviate the senescence of AECs still need to be further explored. In the present study, we observed that bleomycin (BLM) induced enhanced mitophagy accompanied by increased mitochondrial ROS (mito-ROS) content in the murine alveolar epithelial cell line MLE12. While EETs reduced BLM-induced mitophagy and mito-ROS content in MLE12 cells, and the mechanism was related to the regulation of NOX4/Nrf2-mediated redox imbalance. Furthermore, we found that inhibition of EETs degradation could significantly inhibit mitophagy and regulate NOX4/Nrf2 balance to exert anti-oxidant effects in D-galactose-induced premature aging mice. Collectively, these findings may provide new ideas for treating age-related pulmonary diseases by targeting EETs to improve mitochondrial dysfunction and reduce oxidative stress.
Collapse
Affiliation(s)
- Jie-Ru Hong
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Jian-Bing Xiong
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Ping Deng
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Hui Chen
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
13
|
Yu Y, Yang A, He X, Wu B, Wu Y, Li Y, Nie S, Xu B, Wang H, Yu G. Soluble epoxide hydrolase deficiency attenuates airway inflammation in COPD via IRE1α/JNK/AP-1 signaling pathway. J Inflamm (Lond) 2023; 20:36. [PMID: 37915073 PMCID: PMC10621191 DOI: 10.1186/s12950-023-00361-y] [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: 08/22/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Soluble Epoxide Hydrolase (sEH) metabolizes anti-inflammatory epoxyeicosatrienoic acids and critically affects airway inflammation in chronic obstructive pulmonary disease (COPD). Considering the excessive endoplasmic reticulum stress is associated with the earlier onset of COPD. The role of sEH and endoplasmic reticulum stress in the pathogenesis of COPD remains unknown. METHOD 16 weeks of cigarette-exposed mice were used to detect the relationship between sEH and endoplasmic reticulum stress in COPD. Human epithelial cells were used in vitro to determine the regulation mechanism of sEH in endoplasmic reticulum stress induced by cigarette smoke. RESULTS sEH deficiency helps reduce emphysema formation after smoke exposure by alleviating endoplasmic reticulum stress response. sEH deficiency effectively reverses the upregulation of phosphorylation IRE1α and JNK and the nuclear expression of AP-1, alleviating the secretion of inflammatory factors induced by cigarette smoke extract. Furthermore, the treatment with endoplasmic reticulum stress and IRE1α inhibitor downregulated cigarette smoke extract-induced sEH expression and the secretion of inflammatory factors. CONCLUSION sEH probably alleviates airway inflammatory response and endoplasmic reticulum stress via the IRE1α/JNK/AP-1 pathway, which might attenuate lung injury caused by long-term smoking and provide a new pharmacological target for preventing and treating COPD.
Collapse
Affiliation(s)
- Yue Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Ailin Yang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Xin He
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Bo Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Yanjun Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Yunxiao Li
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Shan Nie
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Bo Xu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China.
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China.
| | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China.
| |
Collapse
|
14
|
Yang F, Wendusubilige, Kong J, Zong Y, Wang M, Jing C, Ma Z, Li W, Cao R, Jing S, Gao J, Li W, Wang J. Identifying oxidative stress-related biomarkers in idiopathic pulmonary fibrosis in the context of predictive, preventive, and personalized medicine using integrative omics approaches and machine-learning strategies. EPMA J 2023; 14:417-442. [PMID: 37605652 PMCID: PMC10439879 DOI: 10.1007/s13167-023-00334-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/09/2023] [Indexed: 08/23/2023]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a rare interstitial lung disease with a poor prognosis that currently lacks effective treatment methods. Preventing the acute exacerbation of IPF, identifying the molecular subtypes of patients, providing personalized treatment, and developing individualized drugs are guidelines for predictive, preventive, and personalized medicine (PPPM / 3PM) to promote the development of IPF. Oxidative stress (OS) is an important pathological process of IPF. However, the relationship between the expression levels of oxidative stress-related genes (OSRGs) and clinical indices in patients with IPF is unclear; therefore, it is still a challenge to identify potential beneficiaries of antioxidant therapy. Because PPPM aims to recognize and manage diseases by integrating multiple methods, patient stratification and analysis based on OSRGs and identifying biomarkers can help achieve the above goals. Methods Transcriptome data from 250 IPF patients were divided into training and validation sets. Core OSRGs were identified in the training set and subsequently clustered to identify oxidative stress-related subtypes. The oxidative stress scores, clinical characteristics, and expression levels of senescence-associated secretory phenotypes (SASPs) of different subtypes were compared to identify patients who were sensitive to antioxidant therapy to conduct differential gene functional enrichment analysis and predict potential therapeutic drugs. Diagnostic markers between subtypes were obtained by integrating multiple machine learning methods, their expression levels were tested in rat models with different degrees of pulmonary fibrosis and validation sets, and nomogram models were constructed. CIBERSORT, single-cell RNA sequencing, and immunofluorescence staining were used to explore the effects of OSRGs on the immune microenvironment. Results Core OSRGs classified IPF into two subtypes. Patients classified into subtypes with low oxidative stress levels had better clinical scores, less severe fibrosis, and lower expression of SASP-related molecules. A reliable nomogram model based on five diagnostic markers was constructed, and these markers' expression stability was verified in animal experiments. The number of neutrophils in the immune microenvironment was significantly different between the two subtypes and was closely related to the degree of fibrosis. Conclusion Within the framework of PPPM, this work comprehensively explored the role of OSRGs and their mediated cellular senescence and immune processes in the progress of IPF and assessed their capabilities aspredictors of high oxidative stress and disease progression,targets of the vicious loop between regulated pulmonary fibrosis and OS for targeted secondary and tertiary prevention, andreferences for personalized antioxidant and antifibrotic therapies. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00334-4.
Collapse
Affiliation(s)
- Fan Yang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wendusubilige
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Institute of Ethnic Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jingwei Kong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhan Zong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Manting Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chuanqing Jing
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhaotian Ma
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Institute of Ethnic Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wanyang Li
- Department of Clinical Nutrition, Chinese Academy of Medical Sciences - Peking Union Medical College, Peking Union Medical College Hospital (Dongdan campus), Beijing, China
| | - Renshuang Cao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuwen Jing
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Gao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenxin Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ji Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Hong JR, Jin L, Zhang CY, Zhong WJ, Yang HH, Wang GM, Ma SC, Guan CX, Li Q, Zhou Y. Mitochondrial citrate accumulation triggers senescence of alveolar epithelial cells contributing to pulmonary fibrosis in mice. Heliyon 2023; 9:e17361. [PMID: 37416635 PMCID: PMC10320039 DOI: 10.1016/j.heliyon.2023.e17361] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 07/08/2023] Open
Abstract
Alveolar epithelial cell (AEC) senescence is implicated in the pathogenesis of pulmonary fibrosis (PF). However, the exact mechanism underlying AEC senescence during PF remains poorly understood. Here, we reported an unrecognized mechanism for AEC senescence during PF. We found that, in bleomycin (BLM)-induced PF mice, the expressions of isocitrate dehydrogenase 3α (Idh3α) and citrate carrier (CIC) were significantly down-regulated in the lungs, which could result in mitochondria citrate (citratemt) accumulation in our previous study. Notably, the down-regulation of Idh3α and CIC was related to senescence. The mice with AECs-specific Idh3α and CIC deficiency by adenoviral vector exhibited spontaneous PF and senescence in the lungs. In vitro, co-inhibition of Idh3α and CIC with shRNA or inhibitors triggered the senescence of AECs, indicating that accumulated citratemt triggers AEC senescence. Mechanistically, citratemt accumulation impaired the mitochondrial biogenesis of AECs. In addition, the senescence-associated secretory phenotype from senescent AECs induced by citratemt accumulation activated the proliferation and transdifferentiation of NIH3T3 fibroblasts into myofibroblasts. In conclusion, we show that citratemt accumulation would be a novel target for protection against PF that involves senescence.
Collapse
Affiliation(s)
- Jie-Ru Hong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Guan-Ming Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Sheng-Chao Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- The School of Basic Medical Sciences, Ningxia Medical University Yinchuan 750004, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Qing Li
- Department of Physiology, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
16
|
Zhang CY, Zhong WJ, Liu YB, Duan JX, Jiang N, Yang HH, Ma SC, Jin L, Hong JR, Zhou Y, Guan CX. EETs alleviate alveolar epithelial cell senescence by inhibiting endoplasmic reticulum stress through the Trim25/Keap1/Nrf2 axis. Redox Biol 2023; 63:102765. [PMID: 37269686 DOI: 10.1016/j.redox.2023.102765] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/16/2023] [Accepted: 05/26/2023] [Indexed: 06/05/2023] Open
Abstract
Alveolar epithelial cell (AEC) senescence is a key driver of a variety of chronic lung diseases. It remains a challenge how to alleviate AEC senescence and mitigate disease progression. Our study identified a critical role of epoxyeicosatrienoic acids (EETs), downstream metabolites of arachidonic acid (ARA) by cytochrome p450 (CYP), in alleviating AEC senescence. In vitro, we found that 14,15-EET content was significantly decreased in senescent AECs. Exogenous EETs supplementation, overexpression of CYP2J2, or inhibition of EETs degrading enzyme soluble epoxide hydrolase (sEH) to increase EETs alleviated AECs' senescence. Mechanistically, 14,15-EET promoted the expression of Trim25 to ubiquitinate and degrade Keap1 and promoted Nrf2 to enter the nucleus to exert an anti-oxidant effect, thereby inhibiting endoplasmic reticulum stress (ERS) and alleviating AEC senescence. Furthermore, in D-galactose (D-gal)-induced premature aging mouse model, inhibiting the degradation of EETs by Trifluoromethoxyphenyl propionylpiperidin urea (TPPU, an inhibitor of sEH) significantly inhibited the protein expression of p16, p21, and γH2AX. Meanwhile, TPPU reduced the degree of age-related pulmonary fibrosis in mice. Our study has confirmed that EETs are novel anti-senescence substances for AECs, providing new targets for the treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jia-Xi Duan
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China
| | - Nan Jiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Sheng-Chao Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China; The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Ling Jin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jie-Ru Hong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China.
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
17
|
Zeng J, Liu J, Huang JH, Fu SP, Wang XY, Xi C, Cui YR, Qu F. Aloperine alleviates lipopolysaccharide-induced acute lung injury by inhibiting NLRP3 inflammasome activation. Int Immunopharmacol 2023; 120:110142. [PMID: 37210910 DOI: 10.1016/j.intimp.2023.110142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 05/23/2023]
Abstract
RATIONALE Excessive activation of the NLRP3 inflammasome is involved in the pathological progression of acute lung injury (ALI). Aloperine (Alo) has anti-inflammatory effects in many inflammatory disease models; however, its role in ALI remains elusive. In this study, we addressed the role of Alo in NLRP3 inflammasome activation in both ALI mice and LPS-treated RAW264.7 cells. METHODS The activation of the NLRP3 inflammasome in LPS-induced ALI lungs was investigated in C57BL/6 mice. Alo was administered in order to study its effect on NLRP3 inflammasome activation in ALI. RAW264.7 cells were used to evaluate the underlying mechanism of Alo in the activation of the NLRP3 inflammasome in vitro. RESULTS The activation of the NLRP3 inflammasome occurs in the lungs and RAW264.7 cells under LPS stress. Alo attenuated the pathological injury of lung tissue as well as downregulates the mRNA expression of NLRP3 and pro-caspase-1 in ALI mice and LPS-stressed RAW264.7 cells. The expression of NLRP3, pro-caspase-1, and caspase-1 p10 were also significantly suppressed by Alo in vivo and in vitro. Furthermore, Alo decreased IL-1β and IL-18 release in ALI mice and LPS-induced RAW264.7 cells. In addition, ML385, a Nrf2 inhibitor, weakened the activity of Alo, which inhibited the activation of the NLRP3 inflammasome in vitro. CONCLUSION Alo reduces NLRP3 inflammasome activation via the Nrf2 pathway in ALI mice.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China; Jiangxi Medical College, Shangrao, Jiangxi 334000, China
| | - Jie Liu
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Jun-Hao Huang
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | | | - Xin-Yi Wang
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Chao Xi
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Yan-Ru Cui
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China.
| | - Fei Qu
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China.
| |
Collapse
|
18
|
Zhong WJ, Zhang J, Duan JX, Zhang CY, Ma SC, Li YS, Yang NSY, Yang HH, Xiong JB, Guan CX, Jiang ZX, You ZJ, Zhou Y. TREM-1 triggers necroptosis of macrophages through mTOR-dependent mitochondrial fission during acute lung injury. J Transl Med 2023; 21:179. [PMID: 36879273 PMCID: PMC9990355 DOI: 10.1186/s12967-023-04027-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Necroptosis of macrophages is a necessary element in reinforcing intrapulmonary inflammation during acute lung injury (ALI). However, the molecular mechanism that sparks macrophage necroptosis is still unclear. Triggering receptor expressed on myeloid cells-1 (TREM-1) is a pattern recognition receptor expressed broadly on monocytes/macrophages. The influence of TREM-1 on the destiny of macrophages in ALI requires further investigation. METHODS TREM-1 decoy receptor LR12 was used to evaluate whether the TREM-1 activation induced necroptosis of macrophages in lipopolysaccharide (LPS)-induced ALI in mice. Then we used an agonist anti-TREM-1 Ab (Mab1187) to activate TREM-1 in vitro. Macrophages were treated with GSK872 (a RIPK3 inhibitor), Mdivi-1 (a DRP1 inhibitor), or Rapamycin (an mTOR inhibitor) to investigate whether TREM-1 could induce necroptosis in macrophages, and the mechanism of this process. RESULTS We first observed that the blockade of TREM-1 attenuated alveolar macrophage (AlvMs) necroptosis in mice with LPS-induced ALI. In vitro, TREM-1 activation induced necroptosis of macrophages. mTOR has been previously linked to macrophage polarization and migration. We discovered that mTOR had a previously unrecognized function in modulating TREM-1-mediated mitochondrial fission, mitophagy, and necroptosis. Moreover, TREM-1 activation promoted DRP1Ser616 phosphorylation through mTOR signaling, which in turn caused surplus mitochondrial fission-mediated necroptosis of macrophages, consequently exacerbating ALI. CONCLUSION In this study, we reported that TREM-1 acted as a necroptotic stimulus of AlvMs, fueling inflammation and aggravating ALI. We also provided compelling evidence suggesting that mTOR-dependent mitochondrial fission is the underpinning of TREM-1-triggered necroptosis and inflammation. Therefore, regulation of necroptosis by targeting TREM-1 may provide a new therapeutic target for ALI in the future.
Collapse
Affiliation(s)
- Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Jun Zhang
- Department of Physiology, Hunan University of Medicine, Huaihua, China
| | - Jia-Xi Duan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Sheng-Chao Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.,The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Jian-Bing Xiong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Zhi-Xing Jiang
- Department of Physiology, Hunan University of Medicine, Huaihua, China
| | - Zhi-Jian You
- Department of Anesthesiology, Liuzhou People's Hospital, Liuzhou, China. .,Liuzhou Key Laboratory of Anesthesia and Brain Health, Liuzhou People's Hospital, Liuzhou, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
19
|
Tu M, Wei T, Jia Y, Wang Y, Wu J. Molecular mechanisms of alveolar epithelial cell senescence and idiopathic pulmonary fibrosis: a narrative review. J Thorac Dis 2023; 15:186-203. [PMID: 36794134 PMCID: PMC9922607 DOI: 10.21037/jtd-22-886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 11/25/2022] [Indexed: 12/29/2022]
Abstract
Background and Objective Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial pneumonia of unknown etiology. An increasing number of studies have reported that the incidence of IPF increases with age. Simultaneously, the number of senescent cells increased in IPF. Epithelial cell senescence, an important component of epithelial cell dysfunction, plays a key role in IPF pathogenesis. This article summarizes the molecular mechanisms associated with alveolar epithelial cell senescence and recent advances in the applications of drugs targeting pulmonary epithelial cell senescence to explore novel therapeutic approaches for the treatment of pulmonary fibrosis. Methods All literature published in English on PubMed, Web of Science, and Google Scholar were electronically searched online using the following keyword combinations: aging, alveolar epithelial cell, cell senescence, idiopathic pulmonary fibrosis, WNT/β-catenin, phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt), mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-κB). Key Content and Findings We focused on signaling pathways associated with alveolar epithelial cell senescence in IPF, including WNT/β-catenin, PI3K/Akt, NF-κB, and mTOR signaling pathways. Some of these signaling pathways are involved in alveolar epithelial cell senescence by affecting cell cycle arrest and secretion of senescence-associated secretory phenotype-associated markers. We also found that changes in lipid metabolism in alveolar epithelial cells can be induced by mitochondrial dysfunction, both of which contribute to cellular senescence and development of IPF. Conclusions Decreasing senescent alveolar epithelial cells may be a promising strategy for the treatment of IPF. Therefore, further investigations into new treatments of IPF by applying inhibitors of relevant signaling pathways, as well as senolytic drugs, are warranted.
Collapse
Affiliation(s)
- Mingjin Tu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China;,Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China;,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China;,Peptide and Protein Research and Application Key Laboratory of Guangdong Medical University, Zhanjiang, China
| | - Ting Wei
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China;,Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China;,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China;,Peptide and Protein Research and Application Key Laboratory of Guangdong Medical University, Zhanjiang, China
| | - Yufang Jia
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China;,Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China;,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China;,Peptide and Protein Research and Application Key Laboratory of Guangdong Medical University, Zhanjiang, China
| | - Yajun Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China;,Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China;,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China;,Peptide and Protein Research and Application Key Laboratory of Guangdong Medical University, Zhanjiang, China;,Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Jun Wu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China;,Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China;,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China;,Peptide and Protein Research and Application Key Laboratory of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
20
|
TREM-1 exacerbates bleomycin-induced pulmonary fibrosis by aggravating alveolar epithelial cell senescence in mice. Int Immunopharmacol 2022; 113:109339. [DOI: 10.1016/j.intimp.2022.109339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/30/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
21
|
Vieider L, Zoeller E, Romp E, Schoenthaler M, Hernández-Olmos V, Temml V, Hasenoehrl T, Schuster D, Werz O, Garscha U, Matuszczak B. Synthesis and structure-activity relationships for some novel diflapolin derivatives with benzimidazole subunit. J Enzyme Inhib Med Chem 2022; 37:1752-1764. [PMID: 36124840 PMCID: PMC9518245 DOI: 10.1080/14756366.2022.2087645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/09/2022] [Accepted: 06/05/2022] [Indexed: 11/27/2022] Open
Abstract
A series of derivatives of the potent dual soluble epoxide hydrolase (sEH)/5-lipoxygenase-activating protein (FLAP) inhibitor diflapolin was designed, synthesised, and characterised. These novel compounds, which contain a benzimidazole subunit were evaluated for their inhibitory activity against sEH and FLAP. Molecular modelling tools were applied to analyse structure-activity relationships (SAR) on both targets and to predict solubility and gastrointestinal (GI) absorption. The most promising dual inhibitors of these series are 5a, 6b, and 6c.
Collapse
Affiliation(s)
- Lisa Vieider
- Department of Pharmaceutical Chemistry, Center for Chemistry and Biomedicine, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Eva Zoeller
- Department of Pharmaceutical Chemistry, Center for Chemistry and Biomedicine, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Erik Romp
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University of Jena, Jena, Germany
| | - Martin Schoenthaler
- Department of Pharmaceutical Chemistry, Center for Chemistry and Biomedicine, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Victor Hernández-Olmos
- Branch for Translation Medicine and Pharmacology TMP, Frauenhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt am Main, Germany
| | - Veronika Temml
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Thomas Hasenoehrl
- Department of Pharmaceutical Chemistry, Center for Chemistry and Biomedicine, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - Daniela Schuster
- Department of Pharmaceutical Chemistry, Center for Chemistry and Biomedicine, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University of Jena, Jena, Germany
| | - Ulrike Garscha
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Barbara Matuszczak
- Department of Pharmaceutical Chemistry, Center for Chemistry and Biomedicine, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
22
|
Crosstalk between Depression and Breast Cancer via Hepatic Epoxide Metabolism: A Central Comorbidity Mechanism. Molecules 2022; 27:molecules27217269. [PMID: 36364213 PMCID: PMC9655600 DOI: 10.3390/molecules27217269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Breast cancer (BC) is a serious global challenge, and depression is one of the risk factors and comorbidities of BC. Recently, the research on the comorbidity of BC and depression has focused on the dysfunction of the hypothalamic–pituitary–adrenal axis and the persistent stimulation of the inflammatory response. However, the further mechanisms for comorbidity remain unclear. Epoxide metabolism has been shown to have a regulatory function in the comorbid mechanism with scattered reports. Hence, this article reviews the role of epoxide metabolism in depression and BC. The comprehensive review discloses the imbalance in epoxide metabolism and its downstream effect shared by BC and depression, including overexpression of inflammation, upregulation of toxic diols, and disturbed lipid metabolism. These downstream effects are mainly involved in the construction of the breast malignancy microenvironment through liver regulation. This finding provides new clues on the mechanism of BC and depression comorbidity, suggesting in particular a potential relationship between the liver and BC, and provides potential evidence of comorbidity for subsequent studies on the pathological mechanism.
Collapse
|
23
|
Epoxyeicosatrienoic Acids Inhibit the Activation of Murine Fibroblasts by Blocking the TGF-β1-Smad2/3 Signaling in a PPARγ-Dependent Manner. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7265486. [PMID: 36275905 PMCID: PMC9584742 DOI: 10.1155/2022/7265486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/22/2022] [Indexed: 11/18/2022]
Abstract
Background Epoxyeicosatrienoic acids (EETs), the metabolite of arachidonic acid by cytochrome P450 (CYP), reportedly serve as a vital endogenous protective factor in several chronic diseases. EETs are metabolized by soluble epoxide hydrolase (sEH). We have observed that prophylactic blocking sEH alleviates bleomycin- (BLM-) induced pulmonary fibrosis (PF) in mice. However, the underlying mechanism and therapeutic effects of EETs on PF remain elusive. Objective In this study, we investigated the effect of CYP2J2/EETs on the activation of murine fibroblasts and their mechanisms. Results we found that administration of the sEH inhibitor (TPPU) 7 days after the BLM injection also reversed the morphology changes and collagen deposition in the lungs of BLM-treated mice, attenuating PF. Fibroblast activation is regarded as a critical role of PF. Therefore, we investigated the effects of EETs on the proliferation and differentiation of murine fibroblasts. Results showed that the overexpression of CYP2J2 reduced the cell proliferation and the expressions of α-SMA and PCNA induced by transforming growth factor- (TGF-) β1 in murine fibroblasts. Then, we found that EETs inhibited the proliferation and differentiation of TGF-β1-treated-NIH3T3 cells and primary murine fibroblasts. Mechanistically, we found that 14,15-EET disrupted the phosphorylation of Smad2/3 murine fibroblasts by activating PPARγ, which was completely abolished by a PPARγ inhibitor GW9662. Conclusion our study shows that EETs inhibit the activation of murine fibroblasts by blocking the TGF-β1-Smad2/3 signaling in a PPARγ-dependent manner. Regulating CYP2J2-EET-sEH metabolic pathway may be a potential therapeutic option in PF.
Collapse
|
24
|
Guan XX, Yang HH, Zhong WJ, Duan JX, Zhang CY, Jiang HL, Xiang Y, Zhou Y, Guan CX. Fn14 exacerbates acute lung injury by activating the NLRP3 inflammasome in mice. Mol Med 2022; 28:85. [PMID: 35907805 PMCID: PMC9338586 DOI: 10.1186/s10020-022-00514-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 07/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Uncontrolled inflammation is an important factor in the occurrence and development of acute lung injury (ALI). Fibroblast growth factor-inducible 14 (Fn14), a plasma membrane-anchored receptor, takes part in the pathological process of a variety of acute and chronic inflammatory diseases. However, the role of Fn14 in ALI has not yet been elucidated. This study aimed to investigate whether the activation of Fn14 exacerbated lipopolysaccharide (LPS)-induced ALI in mice. METHODS In vivo, ALI was induced by intratracheal LPS-challenge combined with/without Fn14 receptor blocker aurintricarboxylic acid (ATA) treatment in C57BL/6J mice. Following LPS administration, the survival rate, lung tissue injury, inflammatory cell infiltration, inflammatory factor secretion, oxidative stress, and NLRP3 inflammasome activation were assessed. In vitro, primary murine macrophages were used to evaluate the underlying mechanism by which Fn14 activated the NLRP3 inflammasome. Lentivirus was used to silence Fn14 to observe its effect on the activation of NLRP3 inflammasome in macrophages. RESULTS In this study, we found that Fn14 expression was significantly increased in the lungs of LPS-induced ALI mice. The inhibition of Fn14 with ATA downregulated the protein expression of Fn14 in the lungs and improved the survival rate of mice receiving a lethal dose of LPS. ATA also attenuated lung tissue damage by decreasing the infiltration of macrophages and neutrophils, reducing inflammation, and suppressing oxidative stress. Importantly, we found that ATA strongly inhibited the activation of NLRP3 inflammasome in the lungs of ALI mice. Furthermore, in vitro, TWEAK, a natural ligand of Fn14, amplified the activation of NLRP3 inflammasome in the primary murine macrophage. By contrast, inhibition of Fn14 with shRNA decreased the expression of Fn14, NLRP3, Caspase-1 p10, and Caspase-1 p20, and the production of IL-1β and IL-18. Furthermore, the activation of Fn14 promoted the production of reactive oxygen species and inhibited the activation of Nrf2-HO-1 in activated macrophages. CONCLUSIONS Our study first reports that the activation of Fn14 aggravates ALI by amplifying the activation of NLRP3 inflammasome. Therefore, blocking Fn14 may be a potential way to treat ALI.
Collapse
Affiliation(s)
- Xin-Xin Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
| | - Hui-Ling Jiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China.
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
25
|
COX-2/sEH Dual Inhibitor Alleviates Hepatocyte Senescence in NAFLD Mice by Restoring Autophagy through Sirt1/PI3K/AKT/mTOR. Int J Mol Sci 2022; 23:ijms23158267. [PMID: 35897843 PMCID: PMC9332821 DOI: 10.3390/ijms23158267] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 02/07/2023] Open
Abstract
We previously found that the disorder of soluble epoxide hydrolase (sEH)/cyclooxygenase-2 (COX-2)-mediated arachidonic acid (ARA) metabolism contributes to the pathogenesis of the non-alcoholic fatty liver disease (NAFLD) in mice. However, the exact mechanism has not been elucidated. Accumulating evidence points to the essential role of cellular senescence in NAFLD. Herein, we investigated whether restoring the balance of sEH/COX-2-mediated ARA metabolism attenuated NAFLD via hepatocyte senescence. A promised dual inhibitor of sEH and COX-2, PTUPB, was used in our study to restore the balance of sEH/COX-2-mediated ARA metabolism. In vivo, NAFLD was induced by a high-fat diet (HFD) using C57BL/6J mice. In vitro, mouse hepatocytes (AML12) and mouse hepatic astrocytes (JS1) were used to investigate the effects of PTUPB on palmitic acid (PA)-induced hepatocyte senescence and its mechanism. PTUPB alleviated liver injury, decreased collagen and lipid accumulation, restored glucose tolerance, and reduced hepatic triglyceride levels in HFD-induced NAFLD mice. Importantly, PTUPB significantly reduced the expression of liver senescence-related molecules p16, p53, and p21 in HFD mice. In vitro, the protein levels of γH2AX, p53, p21, COX-2, and sEH were increased in AML12 hepatocytes treated with PA, while Ki67 and PCNA were significantly decreased. PTUPB decreased the lipid content, the number of β-gal positive cells, and the expression of p53, p21, and γH2AX proteins in AML12 cells. Meanwhile, PTUPB reduced the activation of hepatic astrocytes JS1 by slowing the senescence of AML12 cells in a co-culture system. It was further observed that PTUPB enhanced the ratio of autophagy-related protein LC3II/I in AML12 cells, up-regulated the expression of Fundc1 protein, reduced p62 protein, and suppressed hepatocyte senescence. In addition, PTUPB enhanced hepatocyte autophagy by inhibiting the PI3K/AKT/mTOR pathway through Sirt1, contributing to the suppression of senescence. PTUPB inhibits the PI3K/AKT/mTOR pathway through Sirt1, improves autophagy, slows down the senescence of hepatocytes, and alleviates NAFLD.
Collapse
|
26
|
Jiang HL, Yang HH, Liu YB, Zhang CY, Zhong WJ, Guan XX, Jin L, Hong JR, Yang JT, Tan XH, Li Q, Zhou Y, Guan CX. L-OPA1 deficiency aggravates necroptosis of alveolar epithelial cells through impairing mitochondrial function during ALI in mice. J Cell Physiol 2022; 237:3030-3043. [PMID: 35478455 DOI: 10.1002/jcp.30766] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 11/11/2022]
Abstract
Necroptosis, a recently described form of programmed cell death, is the main way of alveolar epithelial cells (AECs) death in acute lung injury (ALI). While the mechanism of how to trigger necroptosis in AECs during ALI has been rarely evaluated. Long optic atrophy protein 1 (L-OPA1) is a crucial mitochondrial inner membrane fusion protein, and its deficiency impairs mitochondrial function. This study aimed to investigate the role of L-OPA1 deficiency-mediated mitochondrial dysfunction in AECs necroptosis. We comprehensively investigated the detailed contribution and molecular mechanism of L-OPA1 deficiency in AECs necroptosis by inhibiting or activating L-OPA1. Firstly, our data showed that L-OPA1 expression was down-regulated in the lungs and AECs under the lipopolysaccharide (LPS) challenge. Furthermore, inhibition of L-OPA1 aggravated the pathological injury, inflammatory response, and necroptosis in the lungs of LPS-induced ALI mice. In vitro, inhibition of L-OPA1 induced necroptosis of AECs, while activation of L-OPA1 alleviated necroptosis of AECs under the LPS challenge. Mechanistically, inhibition of L-OPA1 aggravated necroptosis of AECs by inducing mitochondrial fragmentation and reducing mitochondrial membrane potential. While activation of L-OPA1 had the opposite effects. In summary, these findings indicate for the first time that L-OPA1 deficiency mediates mitochondrial fragmentation, induces necroptosis of AECs, and exacerbates ALI in mice. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hui-Ling Jiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Xin-Xin Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jie-Ru Hong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jin-Tong Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Xiao-Hua Tan
- Experimental Center of Medical Morphology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Qing Li
- Department of Physiology, Hunan University of Medicine, Huaihua, Hunan, 418000, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| |
Collapse
|
27
|
COX-2/sEH Dual Inhibitor PTUPB Attenuates Epithelial-Mesenchymal Transformation of Alveolar Epithelial Cells via Nrf2-Mediated Inhibition of TGF- β1/Smad Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5759626. [PMID: 35509835 PMCID: PMC9060975 DOI: 10.1155/2022/5759626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 04/02/2022] [Accepted: 04/09/2022] [Indexed: 12/15/2022]
Abstract
Background Arachidonic acid (ARA) metabolites are involved in the pathogenesis of epithelial-mesenchymal transformation (EMT). However, the role of ARA metabolism in the progression of EMT during pulmonary fibrosis (PF) has not been fully elucidated. The purpose of this study was to investigate the role of cytochrome P450 oxidase (CYP)/soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2) metabolic disorders of ARA in EMT during PF. Methods A signal intratracheal injection of bleomycin (BLM) was given to induce PF in C57BL/6 J mice. A COX-2/sEH dual inhibitor PTUPB was used to establish the function of CYPs/COX-2 dysregulation to EMT in PF mice. In vitro experiments, murine alveolar epithelial cells (MLE12) and human alveolar epithelial cells (A549) were used to explore the roles and mechanisms of PTUPB on transforming growth factor (TGF)-β1-induced EMT. Results PTUPB treatment reversed the increase of mesenchymal marker molecule α-smooth muscle actin (α-SMA) and the loss of epithelial marker molecule E-cadherin in lung tissue of PF mice. In vitro, COX-2 and sEH protein levels were increased in TGF-β1-treated alveolar epithelial cells (AECs). PTUPB decreased the expression of α-SMA and restored the expression of E-cadherin in TGF-β1-treated AECs, accompanied by reduced migration and collagen synthesis. Moreover, PTUPB attenuated TGF-β1-Smad2/3 pathway activation in AECs via Nrf2 antioxidant cascade. Conclusion PTUPB inhibits EMT in AECs via Nrf2-mediated inhibition of the TGF-β1-Smad2/3 pathway, which holds great promise for the clinical treatment of PF.
Collapse
|
28
|
Zhao Z, Zhang C, Lin J, Zheng L, Li H, Qi X, Huo H, Lou X, Hammock BD, Hwang SH, Bao Y, Luo M. COX-2/sEH Dual Inhibitor PTUPB Alleviates CCl 4 -Induced Liver Fibrosis and Portal Hypertension. Front Med (Lausanne) 2022; 8:761517. [PMID: 35004731 PMCID: PMC8734593 DOI: 10.3389/fmed.2021.761517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/02/2021] [Indexed: 01/02/2023] Open
Abstract
Background: 4-(5-phenyl-3-{3-[3-(4-trifluoromethylphenyl)-ureido]-propyl}-pyrazol-1-yl) -benzenesulfonamide (PTUPB), a dual cyclooxygenase-2 (COX-2)/soluble epoxide hydrolase (sEH) inhibitor, was found to alleviate renal, pulmonary fibrosis and liver injury. However, few is known about the effect of PTUPB on liver cirrhosis. In this study, we aimed to explore the role of PTUPB in liver cirrhosis and portal hypertension (PHT). Method: Rat liver cirrhosis model was established via subcutaneous injection of carbon tetrachloride (CCl4) for 16 weeks. The experimental group received oral administration of PTUPB (10 mg/kg) for 4 weeks. We subsequently analyzed portal pressure (PP), liver fibrosis, inflammation, angiogenesis, and intra- or extrahepatic vascular remodeling. Additionally, network pharmacology was used to investigate the possible mechanisms of PTUPB in live fibrosis. Results: CCl4 exposure induced liver fibrosis, inflammation, angiogenesis, vascular remodeling and PHT, and PTUPB alleviated these changes. PTUPB decreased PP from 17.50 ± 4.65 to 6.37 ± 1.40 mmHg, reduced collagen deposition and profibrotic factor. PTUPB alleviated the inflammation and bile duct proliferation, as indicated by decrease in serum interleukin-6 (IL-6), liver cytokeratin 19 (CK-19), transaminase, and macrophage infiltration. PTUPB also restored vessel wall thickness of superior mesenteric arteries (SMA) and inhibited intra- or extrahepatic angiogenesis and vascular remodeling via vascular endothelial growth factor (VEGF), von Willebrand factor (vWF), etc. Moreover, PTUPB induced sinusoidal vasodilation by upregulating endothelial nitric oxide synthase (eNOS) and GTP-cyclohydrolase 1 (GCH1). In enrichment analysis, PTUPB engaged in multiple biological functions related to cirrhosis, including blood pressure, tissue remodeling, immunological inflammation, macrophage activation, and fibroblast proliferation. Additionally, PTUPB suppressed hepatic expression of sEH, COX-2, and transforming growth factor-β (TGF-β). Conclusion: 4-(5-phenyl-3-{3-[3-(4-trifluoromethylphenyl)-ureido]-propyl}-pyrazol-1-yl)- benzenesulfonamide ameliorated liver fibrosis and PHT by inhibiting fibrotic deposition, inflammation, angiogenesis, sinusoidal, and SMA remodeling. The molecular mechanism may be mediated via the downregulation of the sEH/COX-2/TGF-β.
Collapse
Affiliation(s)
- Zhifeng Zhao
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chihao Zhang
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayun Lin
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zheng
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjie Li
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoliang Qi
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haizhong Huo
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolou Lou
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bruce D Hammock
- Department of Entomology, Nematology and UC Davis Comprehensive Cancer Center, Davis, CA, United States
| | - Sung Hee Hwang
- Department of Entomology, Nematology and UC Davis Comprehensive Cancer Center, Davis, CA, United States
| | - Yongyang Bao
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Luo
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Jankiewicz WK, Barnett SD, Stavniichuk A, Hwang SH, Hammock BD, Belayet JB, Khan AH, Imig JD. Dual sEH/COX-2 Inhibition Using PTUPB-A Promising Approach to Antiangiogenesis-Induced Nephrotoxicity. Front Pharmacol 2021; 12:744776. [PMID: 34955823 PMCID: PMC8695932 DOI: 10.3389/fphar.2021.744776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/21/2021] [Indexed: 01/11/2023] Open
Abstract
Kidney injury from antiangiogenic chemotherapy is a significant clinical challenge, and we currently lack the ability to effectively treat it with pharmacological agents. Thus, we set out to investigate whether simultaneous soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2) inhibition using a dual sEH/COX-2 inhibitor PTUPB could be an effective strategy for treating antiangiogenic therapy-induced kidney damage. We used a multikinase inhibitor, sorafenib, which is known to cause serious renal side effects. The drug was administered to male Sprague-Dawley rats that were on a high-salt diet. Sorafenib was administered over the course of 56 days. The study included three experimental groups; 1) control group (naïve rats), 2) sorafenib group [rats treated with sorafenib only (20 mg/kg/day p.o.)], and 3) sorafenib + PTUPB group (rats treated with sorafenib only for the initial 28 days and subsequently coadministered PTUPB (10 mg/kg/day i.p.) from days 28 through 56). Blood pressure was measured every 2 weeks. After 28 days, sorafenib-treated rats developed hypertension (161 ± 4 mmHg). Over the remainder of the study, sorafenib treatment resulted in a further elevation in blood pressure through day 56 (200 ± 7 mmHg). PTUPB treatment attenuated the sorafenib-induced blood pressure elevation and by day 56, blood pressure was 159 ± 4 mmHg. Urine was collected every 2 weeks for biochemical analysis. After 28 days, sorafenib rats developed pronounced proteinuria (9.7 ± 0.2 P/C), which intensified significantly (35.8 ± 3.5 P/C) by the end of day 56 compared with control (2.6 ± 0.4 P/C). PTUPB mitigated sorafenib-induced proteinuria, and by day 56, it reduced proteinuria by 73%. Plasma and kidney tissues were collected on day 56. Kidney histopathology revealed intratubular cast formation, interstitial fibrosis, glomerular injury, and glomerular nephrin loss at day 56 in sorafenib-treated rats. PTUPB treatment reduced histological features by 30%-70% compared with the sorafenib-treated group and restored glomerular nephrin levels. Furthermore, PTUPB also acted on the glomerular permeability barrier by decreasing angiotensin-II-induced glomerular permeability to albumin. Finally, PTUPB improved in vitro the viability of human mesangial cells. Collectively, our data demonstrate the potential of using PTUPB or dual sEH/COX-2 inhibition as a therapeutic strategy against sorafenib-induced glomerular nephrotoxicity.
Collapse
Affiliation(s)
- Wojciech K. Jankiewicz
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Scott D. Barnett
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anna Stavniichuk
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sung Hee Hwang
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Bruce D. Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Jawad B. Belayet
- Department of Chemistry and Biochemistry, University of Wisconsin Milwaukee, Milwaukee, WI, United States
| | - A. H. Khan
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - John D. Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
30
|
Hudkova O, Krysiuk IP, Kishko TO, Popova NM, Drobot LB, Latyshko NV. Semicarbazide diminishes the signs of bleomycin-induced pulmonary fibrosis in rats. UKRAINIAN BIOCHEMICAL JOURNAL 2021. [DOI: 10.15407/ubj93.05.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
31
|
Khan AH, Hwang SH, Barnett SD, Burkhan A, Jankiewicz WK, Hammock BD, Imig JD. Multitarget molecule, PTUPB, to treat diabetic nephropathy in rats. Br J Pharmacol 2021; 178:4468-4484. [PMID: 34255857 PMCID: PMC8863090 DOI: 10.1111/bph.15623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Diabetic nephropathy is a common complications related to high morbidity and mortality in type 2 diabetes. We investigated the action of the dual modulator, PTUPB, a soluble epoxide hydrolase and cyclooxygenase-2 inhibitor against diabetic nephropathy. EXPERIMENTAL APPROACH Sixteen-week-old type 2 diabetic and proteinuric obese ZSF1 rats were treated with vehicle, PTUPB or enalapril for 8 weeks. Measurements were made of epoxyeicosatrienoic acids, thromboxane B2 (TBX2 ) and prostaglandin E2 (PGE2 ) in the kidney of these and lean ZSF1 rats along with their blood pressure. KEY RESULT Obese ZSF1 rats were diabetic with fivefold higher fasting blood glucose levels and markedly higher HbA1c levels compared with lean ZSF1 rats. PTUPB nor enalapril reduced fasting blood glucose or HbA1c but alleviated the development of diabetic nephropathy. In PTUPB-treated obese ZSF1 rats, glomerular nephrin expression was preserved. Enalapril also alleviated diabetic nephropathy. Diabetic renal injury in obese ZSF1 rats was accompanied by renal inflammation with six to sevenfold higher urinary MCP-1 (CCR2) level and renal infiltration of CD-68 positive cells. PTUPB and enalapril significantly reduced urinary MCP-1 levels and renal mRNA expression of cytokines. Both PTUPB and enalapril lowered blood pressure. PTUPB but not enalapril decreased hyperlipidaemia and liver injury in obese ZSF1 rats. CONCLUSION AND IMPLICATIONS Overall, the dual modulator PTUPB does not treat hyperglycaemia but can effectively alleviate hypertension, diabetic nephropathy, hyperlipidaemia and liver injury in type 2 diabetic rats. Our data further demonstrate that the renal actions of PTUPB are comparable with a current standard diabetic nephropathy treatment.
Collapse
Affiliation(s)
- Abdul Hye Khan
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Scott D. Barnett
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anna Burkhan
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Wojciech K. Jankiewicz
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Bruce D. Hammock
- Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - John D. Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
32
|
Imig JD, Merk D, Proschak E. Multi-Target Drugs for Kidney Diseases. KIDNEY360 2021; 2:1645-1653. [PMID: 35372984 PMCID: PMC8785794 DOI: 10.34067/kid.0003582021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/02/2021] [Indexed: 02/04/2023]
Abstract
Kidney diseases such as AKI, CKD, and GN can lead to dialysis and the need for kidney transplantation. The pathologies for kidney diseases are extremely complex, progress at different rates, and involve several cell types and cell signaling pathways. Complex kidney diseases require therapeutics that can act on multiple targets. In the past 10 years, in silico design of drugs has allowed for multi-target drugs to progress quickly from concept to reality. Several multi-target drugs have been made successfully to target AA pathways and transcription factors for the treatment of inflammatory, fibrotic, and metabolic diseases. Multi-target drugs have also demonstrated great potential to treat diabetic nephropathy and fibrotic kidney disease. These drugs act by decreasing renal TGF-β signaling, inflammation, mitochondrial dysfunction, and oxidative stress. There are several other recently developed multi-target drugs that have yet to be tested for their ability to combat kidney diseases. Overall, there is excellent potential for multi-target drugs that act on several cell types and signaling pathways to treat kidney diseases.
Collapse
Affiliation(s)
- John D. Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| | - Eugen Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
33
|
Duan JX, Guan XX, Yang HH, Mei WX, Chen P, Tao JH, Li Q, Zhou Y. Vasoactive intestinal peptide attenuates bleomycin-induced murine pulmonary fibrosis by inhibiting epithelial-mesenchymal transition: Restoring autophagy in alveolar epithelial cells. Int Immunopharmacol 2021; 101:108211. [PMID: 34634687 DOI: 10.1016/j.intimp.2021.108211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022]
Abstract
Vasoactive intestinal peptide (VIP) is an intrapulmonary neuropeptide with multi-function, including anti-fibrosis. However, the exact role of VIP in pulmonary fibrosis has not been documented. Here, we investigated the protective effect of VIP against pulmonary fibrosis in a murine model induced by bleomycin (BLM). We found that the overexpression of VIP mediated by the adenoviral vector significantly attenuated the lung tissue destruction, reduced the deposition of the extracellular matrix, and inhibited the expression of alpha-smooth muscle actin (α-SMA) in the lungs of mice received BLM. Mechanismly, we found that VIP significantly suppressed the transforming growth factor-beta 1 (TGF-β1)-induced epithelial-mesenchymal transition (EMT) and inhibited the matrix-producing ability of alveolar epithelial cells in vitro. Furthermore, we found that TGF-β1 depressed the autophagy and an autophagy inductor partly reversed the TGF-β1-induced EMT in alveolar epithelial cells. The impaired autophagy was also observed in the lungs of BLM-treated mice, which was restored by VIP treatment. And VIP treatment enhanced autophagy in TGF-β1-stimulated alveolar epithelial cells, contributing to its anti-EMT effect. In summary, our data, for the first time, show that VIP attenuates BLM-induced pulmonary fibrosis in mice with anti-EMT effect through restoring autophagy in alveolar epithelial cells. This study provides a possibility that inhaled long-acting VIP may be an anti-fibrotic drug in the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Xin-Xin Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Wen-Xiu Mei
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Jia-Hao Tao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Qing Li
- Department of Physiology, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
34
|
Epoxyeicosatrienoic Acids and Fibrosis: Recent Insights for the Novel Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms221910714. [PMID: 34639055 PMCID: PMC8509622 DOI: 10.3390/ijms221910714] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/27/2022] Open
Abstract
Organ fibrosis often ends in eventual organ failure and leads to high mortality. Although researchers have identified many effector cells and molecular pathways, there are few effective therapies for fibrosis to date and the underlying mechanism needs to be examined and defined further. Epoxyeicosatrienoic acids (EETs) are endogenous lipid metabolites of arachidonic acid (ARA) synthesized by cytochrome P450 (CYP) epoxygenases. EETs are rapidly metabolized primarily via the soluble epoxide hydrolase (sEH) pathway. The sEH pathway produces dihydroxyeicosatrienoic acids (DHETs), which have lower activity. Stabilized or increased EETs levels exert several protective effects, including pro-angiogenesis, anti-inflammation, anti-apoptosis, and anti-senescence. Currently, intensive investigations are being carried out on their anti-fibrotic effects in the kidney, heart, lung, and liver. The present review provides an update on how the stabilized or increased production of EETs is a reasonable theoretical basis for fibrosis treatment.
Collapse
|
35
|
Kim HS, Moon SJ, Lee SE, Hwang GW, Yoo HJ, Song JW. The arachidonic acid metabolite 11,12-epoxyeicosatrienoic acid alleviates pulmonary fibrosis. Exp Mol Med 2021; 53:864-874. [PMID: 33990688 PMCID: PMC8178404 DOI: 10.1038/s12276-021-00618-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid that are rapidly metabolized into diols by soluble epoxide hydrolase (sEH). sEH inhibition has been shown to increase the biological activity of EETs, which are known to have anti-inflammatory properties. However, the role of EETs in pulmonary fibrosis remains unexplored. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) was used to analyze EETs in the lung tissues of patients with idiopathic pulmonary fibrosis (IPF, n = 29) and controls (n = 15), and the function of 11,12-EET was evaluated in in vitro and in vivo in pulmonary fibrosis models. EET levels in IPF lung tissues, including those of 8,9-EET, 11,12-EET, and 14,15-EET, were significantly lower than those in control tissues. The 11,12-EET/11,12-DHET ratio in human lung tissues also differentiated IPF from control tissues. 11,12-EET significantly decreased transforming growth factor (TGF)-β1-induced expression of α-smooth muscle actin (SMA) and collagen type-I in MRC-5 cells and primary fibroblasts from IPF patients. sEH-specific siRNA and 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU; sEH inhibitor) also decreased TGF-β1-induced expression of α-SMA and collagen type-I in fibroblasts. Moreover, 11,12-EET and TPPU decreased TGF-β1-induced p-Smad2/3 and extracellular-signal-regulated kinase (ERK) expression in primary fibroblasts from patients with IPF and fibronectin expression in Beas-2B cells. TPPU decreased the levels of hydroxyproline in the lungs of bleomycin-induced mice. 11,12-EET or sEH inhibitors could inhibit pulmonary fibrosis by regulating TGF-β1-induced profibrotic signaling, suggesting that 11,12-EET and the regulation of EETs could serve as potential therapeutic targets for IPF treatment. Signaling molecules called eicosanoids, which are derived from fatty acids, can suppress lung damage in idiopathic pulmonary fibrosis (IPF), a chronic, progressive disease in which scar tissue builds up in the lungs, making it hard to breathe. The causes of IPF are unknown. Eicosanoids, which have anti-inflammatory properties, have been studied in various lung diseases. Jin Woo Song at the University of Ulsan College of Medicine in Seoul, South Korea, and co-workers investigated how they might affect IPF. They found that eicosanoid levels were lower in lung tissues from patients with IPF than in healthy tissues. Further investigation showed eicosanoid levels could be boosted by suppressing an enzyme called sEH that degrades them. Thus, suppression of sEH and boosting of eicosanoid levels show promise as therapeutic targets for IPF.
Collapse
Affiliation(s)
- Hak Su Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Su-Jin Moon
- Department of Pulmonary and Critical Care Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Eun Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gi Won Hwang
- Department of Pulmonary and Critical Care Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Song
- Department of Pulmonary and Critical Care Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Cymbopogon winterianus Essential Oil Attenuates Bleomycin-Induced Pulmonary Fibrosis in a Murine Model. Pharmaceutics 2021; 13:pharmaceutics13050679. [PMID: 34065064 PMCID: PMC8150729 DOI: 10.3390/pharmaceutics13050679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 12/26/2022] Open
Abstract
The essential oil of Cymbopogon winterianus (EOCW) is a natural product with antioxidant, anti-inflammatory, and antifibrotic properties. We studied the effect of EOCW in the progression of histological changes of pulmonary fibrosis (PF) in a rodent model. The oil was obtained by hydrodistillation and characterized using gas chromatography–mass spectrometry. Intratracheal instillation of bleomycin was performed in 30 rats to induce PF, while Sham animals were subjected to instillation of saline solution. The treatment was performed using daily oral administration of distilled water, EOCW at 50, 100, and 200 mg/kg, and deflazacort (DFC). After 28 days, hemogram and bronchoalveolar lavage fluid (BALF), tissue levels of malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT) were assayed. Histological grading of PF, immunohistochemical expression of α-smooth muscle actin (α-SMA), and transforming growth factor-β (TGF-β) were also analyzed. The EOCW major compounds were found to be citronellal, geraniol, and citronellol. EOCW significantly reduced inflammation in BALF, reduced MDA levels, and increased SOD activity. EOCW attenuated histological grading of PF and reduced immunohistochemical expression of α-SMA and TGF-β in a dose-dependent way, likely due to the reduction of oxidative stress, inflammation, and TGF-β-induced myofibroblast differentiation.
Collapse
|
37
|
Tiwari S, Yang J, Morisseau C, Durbin-Johnson B, Hammock BD, Gomes AV. Ibuprofen alters epoxide hydrolase activity and epoxy-oxylipin metabolites associated with different metabolic pathways in murine livers. Sci Rep 2021; 11:7042. [PMID: 33782432 PMCID: PMC8007717 DOI: 10.1038/s41598-021-86284-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 03/11/2021] [Indexed: 11/09/2022] Open
Abstract
Over the last decade oxylipins have become more recognized for their involvement in several diseases. Non-steroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen are known to inhibit cyclooxygenase (COX) enzymes, but how NSAIDs affect oxylipins, in addition to COX products, in animal tissues is not well understood. Oxylipins in livers from male and female mice treated with 100 mg/kg/day of ibuprofen for 7 days were investigated. The results showed that ibuprofen treated male livers contained 7 times more altered oxylipins than ibuprofen treated female livers. In male and female livers some prostaglandins were altered, while diols, hydroxy fatty acids and epoxides were significantly altered in male livers. Some soluble epoxide hydrolase (sEH) products, such as 9,10-DiHODE were found to be decreased, while sEH substrates (such as 9(10)-EpODE and 5(6)-EpETrE) were found to be increased in male livers treated with ibuprofen, but not in ibuprofen treated female livers. The enzymatic activities of sEH and microsomal epoxide hydrolase (mEH) were elevated by ibuprofen in both males and females. Analyzing the influence of sex on the effect of ibuprofen on oxylipins and COX products showed that approximately 27% of oxylipins detected were influenced by sex. The results reveal that ibuprofen disturbs not only the COX pathway, but also the CYP450 and lipoxygenase pathways in male mice, suggesting that ibuprofen is likely to generate sex related differences in biologically active oxylipins. Increased sEH activity after ibuprofen treatment is likely to be one of the mechanisms by which the liver reduces the higher levels of EpODEs and EpETrEs.
Collapse
Affiliation(s)
- Shuchita Tiwari
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA, 95616, USA
| | - Jun Yang
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA, 95616, USA
| | - Christophe Morisseau
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA, 95616, USA
| | | | - Bruce D Hammock
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA, 95616, USA
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA, 95616, USA. .,Department of Physiology and Membrane Biology, University of California, Davis, CA, USA.
| |
Collapse
|
38
|
Sun CP, Zhang XY, Morisseau C, Hwang SH, Zhang ZJ, Hammock BD, Ma XC. Discovery of Soluble Epoxide Hydrolase Inhibitors from Chemical Synthesis and Natural Products. J Med Chem 2020; 64:184-215. [PMID: 33369424 DOI: 10.1021/acs.jmedchem.0c01507] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Soluble epoxide hydrolase (sEH) is an α/β hydrolase fold protein and widely distributed in numerous organs including the liver, kidney, and brain. The inhibition of sEH can effectively maintain endogenous epoxyeicosatrienoic acids (EETs) levels and reduce dihydroxyeicosatrienoic acids (DHETs) levels, resulting in therapeutic potentials for cardiovascular, central nervous system, and metabolic diseases. Therefore, since the beginning of this century, the development of sEH inhibitors is a hot research topic. A variety of potent sEH inhibitors have been developed by chemical synthesis or isolated from natural sources. In this review, we mainly summarized the interconnected aspects of sEH with cardiovascular, central nervous system, and metabolic diseases and then focus on representative inhibitors, which would provide some useful guidance for the future development of potential sEH inhibitors.
Collapse
Affiliation(s)
- Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Xin-Yue Zhang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Sung Hee Hwang
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Zhan-Jun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College (Institute) of Integrative Medicine, College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China.,College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou 311121, People's Republic of China
| |
Collapse
|
39
|
p53: A Key Protein That Regulates Pulmonary Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6635794. [PMID: 33312337 PMCID: PMC7721501 DOI: 10.1155/2020/6635794] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/05/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis is a progressively aggravating lethal disease that is a serious public health concern. Although the incidence of this disease is increasing, there is a lack of effective therapies. In recent years, the pathogenesis of pulmonary fibrosis has become a research hotspot. p53 is a tumor suppressor gene with crucial roles in cell cycle, apoptosis, tumorigenesis, and malignant transformation. Previous studies on p53 have predominantly focused on its role in neoplastic disease. Following in-depth investigation, several studies have linked it to pulmonary fibrosis. This review covers the association between p53 and pulmonary fibrosis, with the aim of providing novel ideas to improve the clinical diagnosis, treatment, and prognosis of pulmonary fibrosis.
Collapse
|
40
|
Hiesinger K, Kramer JS, Beyer S, Eckes T, Brunst S, Flauaus C, Wittmann SK, Weizel L, Kaiser A, Kretschmer SBM, George S, Angioni C, Heering J, Geisslinger G, Schubert-Zsilavecz M, Schmidtko A, Pogoryelov D, Pfeilschifter J, Hofmann B, Steinhilber D, Schwalm S, Proschak E. Design, Synthesis, and Structure–Activity Relationship Studies of Dual Inhibitors of Soluble Epoxide Hydrolase and 5-Lipoxygenase. J Med Chem 2020; 63:11498-11521. [DOI: 10.1021/acs.jmedchem.0c00561] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kerstin Hiesinger
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Jan S. Kramer
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Sandra Beyer
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, ZAFES, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| | - Timon Eckes
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, ZAFES, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| | - Steffen Brunst
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Cathrin Flauaus
- Institute of Pharmacology and Clinical Pharmacy, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt a.M., Germany
| | - Sandra K. Wittmann
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Lilia Weizel
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Astrid Kaiser
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Simon B. M. Kretschmer
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Sven George
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Carlo Angioni
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, ZAFES, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| | - Jan Heering
- Branch for Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, ZAFES, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
- Branch for Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| | - Manfred Schubert-Zsilavecz
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Achim Schmidtko
- Institute of Pharmacology and Clinical Pharmacy, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt a.M., Germany
| | - Denys Pogoryelov
- Institute of Biochemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt a.M., Germany
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, ZAFES, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| | - Bettina Hofmann
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
- Branch for Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| | - Stephanie Schwalm
- Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, ZAFES, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9 D-60438 Frankfurt a.M., Germany
- Branch for Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60590 Frankfurt a.M., Germany
| |
Collapse
|
41
|
Zhong WJ, Duan JX, Liu T, Yang HH, Guan XX, Zhang CY, Yang JT, Xiong JB, Zhou Y, Guan CX, Li Q. Activation of NLRP3 inflammasome up-regulates TREM-1 expression in murine macrophages via HMGB1 and IL-18. Int Immunopharmacol 2020; 89:107045. [PMID: 33045564 PMCID: PMC7545267 DOI: 10.1016/j.intimp.2020.107045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022]
Abstract
NLRP3 inflammasome inhibition reduces TREM-1 expression in the lungs of mice with ALI. Activation of NLRP3 inflammasome up-regulates TREM-1 expression in murine macrophages via HMGB1 and IL-18. NLRP3 inflammasome activation induces TREM-1 expression, contributing to the inflammatory network in the lungs of ALI mice.
NOD-, LRR- and pyrin domain-containing 3 (NLRP3) inflammasome and triggering receptor expressed on myeloid cells-1 (TREM-1) are considered critical orchestrators of the inflammatory response in acute lung injury (ALI). However, few assumptions are based on the relationship between them. Here, we investigated the effect of NLRP3 inflammasome activation on the TREM-1 expression in lipopolysaccharide (LPS)-induced ALI and macrophages. We found that inhibition of the NLRP3 inflammasome reduced the TREM-1 expression and pathological lung injury in mice with ALI. Then, primary murine macrophages were used to dissect the underlying mechanistic events of the activation NLRP3 inflammasome involved in the TREM-1 expression. Our results demonstrated that the conditioned medium (CM) from NLRP3 inflammasome-activated-macrophages up-regulated the TREM-1 expression in macrophages, while this effect was reversed by an NLRP3 inflammasome inhibitor MCC950. Furthermore, neutralizing antibodies anti-IL-18 and anti-HMGB1 reduced the TREM-1 expression induced by NLRP3 inflammasome activation. Mechanistically, we found that CM from NLRP3 inflammasome-activated-macrophages increased the level of inhibitor κB kinase protein phosphorylation (p-IκBα) and reactive oxygen species (ROS) content, and promoted IκBα protein degradation in macrophages. While the inhibition of nuclear factor kappa-B (NF-κB) and scavenging ROS eliminated the up-regulation of TREM-1 induced by the NLRP3 inflammasome activation in macrophages. In summary, our study confers NLRP3 inflammasome as a new trigger of TREM-1 signing, which allows additional insight into the pathological of the inflammatory response in ALI.
Collapse
Affiliation(s)
- Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Tian Liu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China; College of Physiology Education, Chongqing University of Arts and Science, Chongqing 412160, China
| | - Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jin-Tong Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Qing Li
- Department of Physiology, Hunan University of Medicine, Huaihua, Hunan 418000, China.
| |
Collapse
|
42
|
Resolution of eicosanoid/cytokine storm prevents carcinogen and inflammation-initiated hepatocellular cancer progression. Proc Natl Acad Sci U S A 2020; 117:21576-21587. [PMID: 32801214 DOI: 10.1073/pnas.2007412117] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Toxic environmental carcinogens promote cancer via genotoxic and nongenotoxic pathways, but nongenetic mechanisms remain poorly characterized. Carcinogen-induced apoptosis may trigger escape from dormancy of microtumors by interfering with inflammation resolution and triggering an endoplasmic reticulum (ER) stress response. While eicosanoid and cytokine storms are well-characterized in infection and inflammation, they are poorly characterized in cancer. Here, we demonstrate that carcinogens, such as aflatoxin B1 (AFB1), induce apoptotic cell death and the resulting cell debris stimulates hepatocellular carcinoma (HCC) tumor growth via an "eicosanoid and cytokine storm." AFB1-generated debris up-regulates cyclooxygenase-2 (COX-2), soluble epoxide hydrolase (sEH), ER stress-response genes including BiP, CHOP, and PDI in macrophages. Thus, selective cytokine or eicosanoid blockade is unlikely to prevent carcinogen-induced cancer progression. Pharmacological abrogation of both the COX-2 and sEH pathways by PTUPB prevented the debris-stimulated eicosanoid and cytokine storm, down-regulated ER stress genes, and promoted macrophage phagocytosis of debris, resulting in suppression of HCC tumor growth. Thus, inflammation resolution via dual COX-2/sEH inhibition is an approach to prevent carcinogen-induced cancer.
Collapse
|
43
|
Li ZM, Xu SY, Feng YZ, Cheng YR, Xiong JB, Zhou Y, Guan CX. The role of NOX4 in pulmonary diseases. J Cell Physiol 2020; 236:1628-1637. [PMID: 32780450 DOI: 10.1002/jcp.30005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/26/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) is a subtype of the NOX family, which is mainly expressed in the pulmonary vasculature and pulmonary endothelial cells in the respiratory system. NOX4 has unique characteristics, and is a constitutively active enzyme that primarily produces hydrogen peroxide. The signaling pathways associated with NOX4 are complicated. Negative and positive feedback play significant roles in regulating NOX4 expression. The role of NOX4 is controversial because NOX4 plays a protective or damaging role in different respiratory diseases. This review summarizes the structure, enzymatic properties, regulation, and signaling pathways of NOX4. This review then introduces the roles of NOX4 in different diseases in the respiratory system, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, and pulmonary fibrosis.
Collapse
Affiliation(s)
- Zi-Ming Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Sheng-Ya Xu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yi-Zhuo Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu-Rui Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
44
|
Luo XQ, Duan JX, Yang HH, Zhang CY, Sun CC, Guan XX, Xiong JB, Zu C, Tao JH, Zhou Y, Guan CX. Epoxyeicosatrienoic acids inhibit the activation of NLRP3 inflammasome in murine macrophages. J Cell Physiol 2020; 235:9910-9921. [PMID: 32452554 DOI: 10.1002/jcp.29806] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/09/2020] [Indexed: 12/17/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) derived from arachidonic acid exert anti-inflammation effects. We have reported that blocking the degradation of EETs with a soluble epoxide hydrolase (sEH) inhibitor protects mice from lipopolysaccharide (LPS)-induced acute lung injury (ALI). The underlying mechanisms remain essential questions. In this study, we investigated the effects of EETs on the activation of nucleotide-binding domain leucine-rich repeat-containing receptor, pyrin domain-containing-3 (NLRP3) inflammasome in murine macrophages. In an LPS-induced ALI murine model, we found that sEH inhibitor 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl), TPPU, profoundly attenuated the pathological injury and inhibited the activation of the NLRP3 inflammasome, characterized by the reduction of the protein expression of NLRP3, ASC, pro-caspase-1, interleukin precursor (pro-IL-1β), and IL-1β p17 in the lungs of LPS-treated mice. In vitro, primary peritoneal macrophages from C57BL/6 were primed with LPS and activated with exogenous adenosine triphosphate (ATP). TPPU treatment remarkably reduced the expression of NLRP3 inflammasome-related molecules and blocked the activation of NLRP3 inflammasome. Importantly, four EETs (5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET) inhibited the activation of NLRP3 inflammasome induced by LPS + ATP or LPS + nigericin in macrophages in various degree. While the inhibitory effect of 5,6-EET was the weakest. Mechanismly, EETs profoundly decreased the content of reactive oxygen species (ROS) and restored the calcium overload in macrophages receiving LPS + ATP stimulation. In conclusion, this study suggests that EETs inhibit the activation of the NLRP3 inflammasome by suppressing calcium overload and ROS production in macrophages, contributing to the therapeutic potency to ALI.
Collapse
Affiliation(s)
- Xiao-Qin Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Basic Medicine, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China.,Department of Medical Technology, Changsha Health Vocational College, Changsha, Hunan, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China
| | - Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cheng Zu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jia-Hao Tao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
45
|
Yang HH, Duan JX, Liu SK, Xiong JB, Guan XX, Zhong WJ, Sun CC, Zhang CY, Luo XQ, Zhang YF, Chen P, Hammock BD, Hwang SH, Jiang JX, Zhou Y, Guan CX. A COX-2/sEH dual inhibitor PTUPB alleviates lipopolysaccharide-induced acute lung injury in mice by inhibiting NLRP3 inflammasome activation. Theranostics 2020; 10:4749-4761. [PMID: 32308747 PMCID: PMC7163435 DOI: 10.7150/thno.43108] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/08/2020] [Indexed: 01/11/2023] Open
Abstract
Rationale: Dysregulation of arachidonic acid (ARA) metabolism results in inflammation; however, its role in acute lung injury (ALI) remains elusive. In this study, we addressed the role of dysregulated ARA metabolism in cytochromes P450 (CYPs) /cyclooxygenase-2 (COX-2) pathways in the pathogenesis of lipopolysaccharide (LPS)-induced ALI in mice. Methods: The metabolism of CYPs/COX-2-derived ARA in the lungs of LPS-induced ALI was investigated in C57BL/6 mice. The COX-2/sEH dual inhibitor PTUPB was used to establish the function of CYPs/COX-2 dysregulation in ALI. Primary murine macrophages were used to evaluate the underlying mechanism of PTUPB involved in the activation of NLRP3 inflammasome in vitro. Results: Dysregulation of CYPs/COX-2 metabolism of ARA occurred in the lungs and in primary macrophages under the LPS challenge. Decrease mRNA expression of Cyp2j9, Cyp2j6, and Cyp2j5 was observed, which metabolize ARA into epoxyeicosatrienoic acids (EETs). The expressions of COX-2 and soluble epoxide hydrolase (sEH), on the other hand, was significantly upregulated. Pre-treatment with the dual COX-2 and sEH inhibitor, PTUPB, attenuated the pathological injury of lung tissues and reduced the infiltration of inflammatory cells. Furthermore, PTUPB decreased the pro-inflammatory factors, oxidative stress, and activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in LPS-induced ALI mice. PTUPB pre-treatment remarkably reduced the activation of macrophages and NLRP3 inflammasome in vitro. Significantly, both preventive and therapeutic treatment with PTUPB improved the survival rate of mice receiving a lethal dose of LPS. Conclusion: The dysregulation of CYPs/COX-2 metabolized ARA contributes to the uncontrolled inflammatory response in ALI. The dual COX-2 and sEH inhibitor PTUPB exerts anti-inflammatory effects in treating ALI by inhibiting the NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Shao-Kun Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xiao-Qin Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yan-Feng Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jian-Xin Jiang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
46
|
PTUPB ameliorates high-fat diet-induced non-alcoholic fatty liver disease via inhibiting NLRP3 inflammasome activation in mice. Biochem Biophys Res Commun 2020; 523:1020-1026. [PMID: 31973813 DOI: 10.1016/j.bbrc.2019.12.131] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/31/2019] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects 25% of the global adult population, and no effective pharmacological treatment has been found. Products of arachidonic acid metabolism have been developed into a novel therapy for metabolic syndrome and diabetes. It has been demonstrated that protective actions of a novel dual cyclooxygenase-2 (COX-2) and soluble epoxide hydrolase (sEH) inhibitor, PTUPB, on the metabolic abnormalities. Here, we investigated the effects of PTUPB on hepatic steatosis in high-fat diet (HFD)-induced obese mice, as well as in hepatocytes in vitro. We found that PTUPB treatment reduced body weight, liver weight, liver triglyceride and cholesterol content, and the expression of lipolytic/lipogenic and lipid uptake related genes (Acc, Cd36, and Cidec) in HFD mice. In addition, PTUPB treatment arrested fibrotic progression with a decrease of collagen deposition and expression of Col1a1, Col1a3, and α-SMA. In vitro, PTUPB decreased palmitic acid-induced lipid deposition and downregulation of lipolytic/lipogenic genes (Acc and Cd36) in hepatocytes. Additionally, we found that PTUPB reduced the production of pro-inflammatory cytokines and suppressed the NLRP3 inflammasome activation in HFD mice and hepatocytes. In conclusion, dual inhibition of COX-2/sEH attenuates hepatic steatosis by inhibiting the NLRP3 inflammasome activation. PTUPB might be a promising potential therapy for liver steatosis associated with obesity.
Collapse
|