1
|
Li S, Gong WL, Liu L, Shao B, Jiang SL, Li H, Song Y, Han GZ, Zhang ZQ. SiO 2 particles induce pulmonary fibrosis by modulating NLRP3 through the ROS/Keap1/Nrf2 signaling pathway in rats. Food Chem Toxicol 2025; 202:115520. [PMID: 40334973 DOI: 10.1016/j.fct.2025.115520] [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: 03/19/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/09/2025]
Abstract
Recent studies have shown that the activation of the ROS-dependent NLRP3 inflammasome plays a key role in the pathogenesis of silicosis; however, the mechanism by which SiO2-induced ROS activates NLRP3 remains unclear. In this study, rats were intratracheally instilled with a SiO2 suspension once and then received daily intravenous injections of NAC (at doses of 20, 40, and 80 mg/kg, respectively) to inhibit SiO2-induced ROS. Rats that were intratracheally instilled with a SiO2 suspension once served as silicosis models, while those that were intratracheally instilled with PBS once served as controls. After 40 days, lung samples were taken for pathological observation, and the BALF was collected to measure ROS levels. The mRNA and protein expression levels of Keap1/Nrf2 signaling indicators (Keap1, Nrf2) and NLRP3 inflammasome indicators (NLRP3, GSDMD) were detected. The results showed that the Keap1/Nrf2 signaling pathway and the NLRP3 were activated in the silicosis rat lungs, accompanied by an increase in ROS levels. When ROS was inhibited, the Keap1/Nrf2 signaling pathway, the NLRP3, and the degree of pulmonary fibrosis were all suppressed in a dose-dependent manner. Therefore, we conclude that SiO2 particles induce pulmonary fibrosis in rats by modulating the NLRP3 inflammasome via the ROS/Keap1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Shuang Li
- Department of Public Health, Jining Medical University, Jining, China; Shandong Weixuankang Technology Innovation Co. LTD, Tai An, China
| | - Wei-Lei Gong
- Department of Public Health, Jining Medical University, Jining, China
| | - Lin Liu
- Health Management Center of Affiliated Hospital of Jining Medical University, Jining, China
| | - Bo Shao
- Department of Public Health, Jining Medical University, Jining, China
| | - Shun-Li Jiang
- Department of Public Health, Jining Medical University, Jining, China
| | - Huan Li
- Department of Public Health, Jining Medical University, Jining, China
| | - Ye Song
- Department of Public Health, Jining Medical University, Jining, China
| | - Gui-Zhi Han
- Department of Public Health, Jining Medical University, Jining, China.
| | - Zhao-Qiang Zhang
- Department of Public Health, Jining Medical University, Jining, China.
| |
Collapse
|
2
|
Zheng C, Zhang L, Sun Y, Ma Y, Zhang Y. Alveolar epithelial cell dysfunction and epithelial-mesenchymal transition in pulmonary fibrosis pathogenesis. Front Mol Biosci 2025; 12:1564176. [PMID: 40343260 PMCID: PMC12058482 DOI: 10.3389/fmolb.2025.1564176] [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: 01/21/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Pulmonary fibrosis (PF) is a progressive and lethal interstitial lung disease characterized by aberrant scar formation and destruction of alveolar architecture. Dysfunctional alveolar epithelial cells (AECs) play a central role in initiating PF, where chronic injury triggers apoptosis and disrupts epithelial homeostasis, leading to epithelial-mesenchymal transition (EMT). This dynamic reprogramming process causes AECs to shed epithelial markers and adopt a mesenchymal phenotype, fueling fibroblast activation and pathological extracellular matrix (ECM) deposition. This review systematically explores the multi-layered mechanisms driving AECs dysfunction and EMT, focusing on core signaling axes such as transforming growth factor-β (TGF-β)/Smad, WNT/β-catenin, NF-κB-BRD4, and nuclear factor erythroid 2-related factor 2 (Nrf2), which regulate EMT and fibroblast-ECM interactions. It also highlights emerging regulators, including metabolic reprogramming, exosomal miRNA trafficking, and immune-epithelial interactions. Furthermore, understanding these mechanisms is essential for developing targeted therapeutic strategies to modulate these pathways and halt or reverse fibrosis progression, offering critical insights into potential clinical treatments for PF.
Collapse
Affiliation(s)
- Caopei Zheng
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yuqing Sun
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| |
Collapse
|
3
|
Wang X, Wang Q, Zhou P, Zhang J, Su H, Liu F, Wu J, Xiao F, Liu L, Han L. Rhoifolin improves bleomycin-induced fibrosis in vivo and cell damage in vitro both related to NRF2/HO-1 pathway. BMC Pulm Med 2025; 25:117. [PMID: 40087614 PMCID: PMC11907914 DOI: 10.1186/s12890-025-03526-y] [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: 11/02/2023] [Accepted: 01/24/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a chronic interstitial lung illness characterized by its high worldwide prevalence, unknown etiology, and dismal prognosis. Lonicerae Japonicae Flos, a commonly used traditional Chinese medicine for treating PF, is rich in Rhoifolin (ROF). Although numerous studies have demonstrated the anti-inflammatory properties of ROF, its potential anti-fibrotic effects remain uncertain. METHODS In this study, we established a PF model in Sprague-Dawley (SD) rats utilizing Bleomycin (BLM). We then assessed the impact of ROF on lung histology and appearance, measured the content level of Superoxide dismutase (SOD) in rat serum, and analyzed changes in α-SMA, TGF-β relative mRNA expression using PCR, measured SMAD Family Member 7 (Smad7), and Heme Oxygenase-1 (HO-1) protein expression in rat lung tissue by Western Blot. Additionally, we induced a cell injury model in A549 cells with BLM. Then after ROF administration, we detected the cell viability by MTT assay, measured N-cadherin, α-SMA, and Vimentin mRNA levels via real-time PCR, and analyzed the expression changes of N-cadherin, Nuclear factor erythroid 2-related factor 2(Nrf2), HO-1, Smad7 proteins by Western Blot. RESULTS The results indicated that ROF mitigated lung tissue damage and reduced the degree of PF in the lung tissue of rats with PF. Furthermore, In vivo, ROF reduced the expression of N-cadherin protein while increasing the expression of Smad7, and HO-1 proteins and decreasing the relative mRNA expression of α-SMA and TGF-β, and increased the expresson of SOD in rat serum In vitro, cell injury was induced in A549 cells using BLM. After ROF administration, the relative mRNA expression of α-SMA, N-cadherin, and Vimentin decreased significantly, and the protein expression of N-cadherin decreased, while the protein expression of Nrf2, HO-1, and Smad7 increased significantly. CONCLUSION This study demonstrates that ROF can mitigate the symptoms of PF to a certain degree, and its mechanism of action is intimately linked to the Nrf2/HO-1 signaling pathway. Therefore, this study indicates that ROF may serve as a potential therapeutic agent for treating PF. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Xuehua Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- College of Traditional Chinese Medicine, Zhanjiang University of Science and Technology, Zhanjiang, Guangdong, 524094, China
| | - Qing Wang
- Chongqing Traditional Chinese Medicine Research Institute, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400011, China
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510006, PR China
| | - Panyu Zhou
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510006, PR China
| | - Junhong Zhang
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510006, PR China
| | - Haojie Su
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510006, PR China
| | - Fanlu Liu
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510006, PR China
| | - Jingjing Wu
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510006, PR China
| | - Fengxia Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Liang Liu
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510006, PR China.
| | - Ling Han
- Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510006, PR China.
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P. R. China.
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, P. R. China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China.
| |
Collapse
|
4
|
Al-Gholam MAS, Abd-Elhafiz HI, Tayel SG. Effect of Alpinia officinarum Rhizome extract on experimentally induced lung fibrosis: The pertinent role of Sirt1 and Nrf2 antioxidant pathways. Morphologie 2025; 109:100940. [PMID: 39694016 DOI: 10.1016/j.morpho.2024.100940] [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: 06/29/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a frequently reported COVID-19 sequela. It is a progressive disorder characterized by respiratory failure and death. The properties of Alpinia officinarum Rhizomes (AO) make it a highly potent antioxidant, anti-inflammatory, and antifibrotic agent. This study has evaluated AO's protective effects on bleomycin-induced PF in rats and investigated the underlying mechanisms. MATERIAL AND METHODS Bleomycin (5mg/kg, intratracheally) was used to induce PF in albino rats, and then, AO extract (200mg/kg/daily, orally) was administrated for 28days post-bleomycin-instillation. After euthanizing the rats, the biochemical, quantitative real-time polymerase chain reaction (qPCR) and histopathological examination of lung tissue were determined. RESULTS Findings have revealed that bleomycin significantly increased the tissue level of malondialdehyde, tumor necrosis factor-alpha, and interleukin-6, Silent information regulator 1 (Sirt1), and nuclear factor erythroid 2-related factor 2 (Nrf2) mRNA levels. Furthermore, the total antioxidant capacity level decreased in the lungs of bleomycin-instilled rats. However, AO extract significantly decreased histopathological injuries in hematoxylin & eosin, Masson's trichrome-stained sections, inducible nitric oxide synthase and α-smooth muscle actin, transforming growth factor beta 1 immunoexpression. CONCLUSION Alpinia officinarum Rhizomes extract appears to protect against bleomycin-induced PF, possibly due to its antioxidant, anti-inflammatory, and antifibrotic properties.
Collapse
Affiliation(s)
- Marwa A S Al-Gholam
- Anatomy and Embryology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia, Egypt.
| | - Huda I Abd-Elhafiz
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia, Egypt.
| | - Sara G Tayel
- Anatomy and Embryology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia, Egypt.
| |
Collapse
|
5
|
Yang JJ, Liu HJ, Wang YX, Wang LP, Gu JJ, Gao JY, Ren KQ, Min LF. Oxidative Stress and Epithelial-Mesenchymal Transition: The Impact of Ubiquitin C-terminal Hydrolase L1 in Cigarette Smoke-Induced COPD. Lung 2025; 203:36. [PMID: 40000498 DOI: 10.1007/s00408-025-00790-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/06/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE Cigarette smoke (CS) has been demonstrated to mediate oxidative stress (OS) and epithelial-mesenchymal transition (EMT) in bronchial epithelial cells, thereby contributing to airway remodeling in chronic obstructive pulmonary disease (COPD). Studies have shown upregulation of Ubiquitin C-terminal hydrolase L1 (UCHL1), a deubiquitinating enzyme, in the airway epithelium of smokers. Many studies indicate that UCHL1's regulation of EMT and OS has a complex role in various cell types, including respiratory epithelium. Thus, we aimed to investigate UCHL1's regulation of EMT, OS, and related mechanisms in cigarette smoke-exposed airway epithelium. METHODS Exposure to cigarette smoke (CS) or cigarette smoke extract (CSE) was employed to establish both animal and cellular models. Protein expression was analyzed using immunohistochemistry, immunofluorescence, and Western blotting. Lentiviral UCHL1 or GPX1-siRNA was used to modulate UCHL1 or GPX1 expression, respectively. Transwell assays were employed to evaluate cell migration and EMT-related alterations. Oxidative stress levels were assessed using specific assay kits. RESULTS This study validated that exposure to CS induces UCHL1 expression in bronchial epithelial cells both in vitro and in vivo, a phenomenon positively correlated with increased OS and EMT in the airway. Notably, UCHL1 overexpression counteracted CSE's impact on EMT markers, cell migration, and oxidative stress in BEAS-2B cells, while UCHL1 knockdown exacerbated these effects. Furthermore, in BEAS-2B cells treated with CSE, upregulation of UCHL1 was found to enhance the expression of glutathione peroxidase 1 (GPX1), an antioxidant enzyme. The effect of UCHL1 overexpression on EMT-related protein markers and cell migration was reversed upon GPX1 silencing via siRNA. CONCLUSIONS These findings suggest that UCHL1-mediated regulation of GPX1 expression alleviates cigarette smoke-induced EMT-related protein markers change and cell migration in BEAS-2B cell.
Collapse
Affiliation(s)
- Jing Jing Yang
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hong Jun Liu
- Department of Respiratory Medicine, Jingjiang People's Hospital Affiliated to Yangzhou University, Taizhou, Jiangsu, China
| | - Yu Xiu Wang
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Li Ping Wang
- Biospecimen Library, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jian Jun Gu
- Department of Cardiology, Institute of Translational Medicine, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jun Yin Gao
- Pulmonary and Critical Care Medicine, Yancheng No.1 People's Hospital, 224000, Yancheng, Jiangsu, China
| | - Kai Qi Ren
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ling Feng Min
- Department of Pulmonary and Critical Care Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
6
|
Zhao L, Li J, Dang Y, Fisher D, Hien NTT, Musabaev E, Pronyuk K, Zhao L. Protective role of sulforaphane in lipid metabolism-related diseases. Mol Biol Rep 2025; 52:241. [PMID: 39961997 DOI: 10.1007/s11033-025-10358-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/11/2025] [Indexed: 05/09/2025]
Abstract
Sulforaphane (SFN) is a phytochemical bioactive substance commonly found in cruciferous plants, such as broccoli and mustard. It has been reported to possess antibacterial, anti-inflammatory, anti-oxidant, anti-cancer and autophagy regulating properties. Recent studies have revealed that SFN regulates fat metabolism both in vivo and in vitro through various mechanisms, including alleviating endoplasmic reticulum stress, inhibiting inflammatory response and improving mitochondrial dysfunction, involving Nrf2/ARE, NF-κB, NLRP3 inflammasome, HDAC8-PGC1α axis and other signaling pathways. By curbing complications associated with abnormal fat metabolic diseases, SFN exhibits therapeutic effects on conditions like obesity, fatty liver disease, atherosclerosis, type 2 diabetes, etc., with minimal side effects. Therefore, it holds promise as a potential alternative treatment for lipid metabolism-related diseases. Although its extraction method has been matured, the thermal instability and preservation difficulties of SFN limit its clinical promotion. More effective and low-cost methods to improve the stability and production of SFN remain to be further studied. This paper reviews the physiological and biological activities of SFN, and summarizes the protective effects and molecular mechanisms of SFN in diseases related to abnormal lipid metabolism. Additionally, it proposes potential challenges, possible solutions and future research directions in the clinical application of SFN.
Collapse
Affiliation(s)
- Lingfeng Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahuan Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Cape Town, 7100, South Africa
| | | | - Erkin Musabaev
- The Research Institute of Virology, Ministry of Health, 100122, Tashkent, Uzbekistan
| | - Khrystyna Pronyuk
- Infectious Diseases Department, O.Bogomolets National Medical University, Kiev, 02132, Ukraine
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
7
|
Zeng L, Wang Y, Huang Y, Yang W, Zhou P, Wan Y, Tao K, Li R. IRG1/itaconate enhances efferocytosis by activating Nrf2-TIM4 signaling pathway to alleviate con A induced autoimmune liver injury. Cell Commun Signal 2025; 23:63. [PMID: 39910615 PMCID: PMC11796036 DOI: 10.1186/s12964-025-02075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
Immune response gene 1 (IRG1) is highly expressed in mitochondria of macrophages in a pro-inflammatory state. IRG1 and its metabolites play important roles in infection, immune-related diseases and tumor progression by exerting resistance of pathogens, attenuating inflammation and producing antioxidant substances through various pathways and mechanisms. IRG1 deficiency aggravates liver injury. Efferocytosis is a vital mechanism for preventing the progression of inflammatory tissue damage. However, the mechanism by how IRG1/itaconate regulates efferocytosis in autoimmune hepatitis has yet to be fully understood. Therefore, we explored the influence of IRG1-/- on efferocytosis and its effects on regulating the nuclear factor erythroid 2-associated factor 2 (Nrf2)-T-cell immunoglobulin domain and mucin domain 4 (TIM4) pathway and autoimmune liver injury. An autoimmune hepatitis model was established by injecting Con A into wild-type and IRG1-/- mice via the tail vein. Liver injury and inflammatory response were assessed. The efferocytosis role of IRG1-/- macrophages and its potential regulatory mechanisms were also analysed. Exogenous 4-octyl itaconate (OI) supplementation promoted the expression of Nrf2 and TIM4 and restored IRG1-/- bone marrow-derived macrophage (BMDM) efferocytosis, whereas inhibition of Nrf2 mediated by ML385 led to impaired efferocytosis of BMDMs, decreased expression of TIM4, and aggravated liver inflammation injury. Additionally, after supplementing Nrf2-/- BMDMs with exogenous OI, we evaluated the changes in its efferocytosis effect, efferocytosis did not change, and the protective effect of OI disappeared. However, when TIM4 was blocked, the efferocytotic effect of BMDMs was attenuated, inflammatory liver injury and oxidative stress were aggravated. OI promoted the transformation of macrophages into M2 macrophages, and this was inhibited when TIM4 was blocked. To our best understanding, this is the initial exploration to show that TIM4, a downstream molecule of the IRG1/itaconate-Nrf2 pathway, regulates macrophage efferocytosis. These findings suggest a new mechanism and potential treatment for promoting the resolution of inflammation and efferocytosis in autoimmune hepatitis.
Collapse
Affiliation(s)
- Liwu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Yaxin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yongzhou Huang
- Department of General Surgery, First Affiliated Hospital of Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Wenchang Yang
- Department of Gastroenterology Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Pei Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Yaqi Wan
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China.
| | - Ruidong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
8
|
Lu C, Liu Y, Ren F, Zhang H, Hou Y, Zhang H, Chen Z, Du X. HO-1: An emerging target in fibrosis. J Cell Physiol 2025; 240:e31465. [PMID: 39420552 DOI: 10.1002/jcp.31465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/08/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Fibrosis, an aberrant reparative response to tissue injury, involves a disruption in the equilibrium between the synthesis and degradation of the extracellular matrix, leading to its excessive accumulation within normal tissues, and culminating in organ dysfunction. Manifesting in the terminal stages of nearly all chronic ailments, fibrosis carries a high mortality rate and poses a significant threat to human health. Heme oxygenase-1 (HO-1) emerges as an endogenous protective agent, mitigating tissue damage through its antioxidant, anti-inflammatory, and antiapoptotic properties. Numerous studies have corroborated HO-1's potential as a therapeutic target in anti-fibrosis treatment. This review delves into the structural and functional attributes, and the upstream and downstream pathways of HO-1. Additionally, the regulatory networks and mechanisms of HO-1 in cells associated with fibrosis are elucidated. The role of HO-1 in various fibrosis-related diseases is also explored. Collectively, this comprehensive information serves as a foundation for future research and augments the viability of HO-1 as a therapeutic target for fibrosis.
Collapse
Affiliation(s)
- Chenxi Lu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Yuan Liu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Feifei Ren
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Haoran Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Yafang Hou
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Hong Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Zhiyong Chen
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Xia Du
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| |
Collapse
|
9
|
Li K, Liu H, Li M, Sun M, Peng X, Wu Y, Tian Y, Liu X, Li J. Mechanistic insights into the treatment of pulmonary fibrosis with bioactive components from traditional chinese medicine via matrix stiffness-mediated EMT. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156266. [PMID: 39580995 DOI: 10.1016/j.phymed.2024.156266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease with limited therapeutic options. Our previous research has shown that the Jinshui Huanxian formula (JHF) is effective in treating IPF. However, the biomechanical mechanisms of its refined components, known as the effective-component compatibility of JHF II (ECC-JHF II), are not well understood. PURPOSE This study aims to explore how bioactive components from traditional Chinese medicine (TCM) impact the biomechanical progression of pulmonary fibrosis. STUDY DESIGN AND METHODS A mouse model of pulmonary fibrosis was established by a single intratracheal instillation of bleomycin (Bleomycin). Pulmonary function, pathological changes, collagen deposition, lung tissue stiffness, and EMT markers were evaluated at the end of the study. Polyethylene glycol hydrogels with adjustable stiffness were used to mimic both normal and pathological lung conditions. The effects of ECC-JHF II on matrix stiffness-mediated EMT were assessed by quantitative real-time PCR, western blot, and immunofluorescence. The biomechanical mechanisms underlying ECC-JHF II on EMT and pulmonary fibrosis were verified both in vivo and in vitro. RESULTS ECC-JHF II significantly improved bleomycin (Bleomycin)-induced pulmonary fibrosis in mice, manifested as increased tidal volume and 50 % tidal volume expiratory flow, reduced lung tissue stiffness, and decreased EMT markers. Histopathological analysis showed reduced inflammation, alveolar damage, and collagen deposition. In vitro, ECC-JHF II reversed the EMT phenotypic transition induced by substrate stiffness, demonstrated by the upregulation of E-cadherin, occludin, and zonula occluden-1, and the downregulation of N-cadherin, vimentin, caldesmon 1 and tropomyosin 1. Moreover, ECC-JHF II could inhibit integrin/ROCK/MRTF signaling in vitro and in vivo. Silencing integrin β1 or activating it with pyrintegrin further confirmed the role of integrin β1 in the mechanotransduction pathway and the efficacy of ECC-JHF II. CONCLUSION Taken together, the findings of this study indicate that ECC-JHF II exerts a therapeutic effect on pulmonary fibrosis through the attenuation of lung tissue stiffness and inhibition of EMT, potentially via the integrin/ROCK/MRTF signaling pathway.
Collapse
Affiliation(s)
- Kangchen Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Han Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Mingyan Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Meihao Sun
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xiling Peng
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Yuanyuan Wu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Yange Tian
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xinguang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| |
Collapse
|
10
|
Zheng Q, Lei FP, Hui S, Tong M, Liang LH. Ginsenoside Rb1 Relieves Cellular Senescence and Pulmonary Fibrosis by Promoting NRF2/QKI/SMAD7 Axis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2491-2509. [PMID: 39756830 DOI: 10.1142/s0192415x24500952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Cellular senescence is an adverse factor in the development of pulmonary fibrosis (PF). Ginsenoside Rb1 has been found to inhibit both cellular senescence and PF. This study aimed to elucidate the molecular mechanisms by which ginsenoside Rb1 regulates cellular senescence and PF. A PF mouse model was established by Bleomycin (BLM) administration, and a cell model of senescence was constructed using MRC-5 cells treated with Adriamycin RD (ARD) administration. Hematoxylin and Eosin (HE) staining and Masson staining were employed to evaluate cellular structure and collagen fiber content. RT-qPCR and western blotting were used to detect mRNA and protein expression of the target genes. Enzyme-linked Immunosorbent Assay (ELISA) was applied to measure the protein concentration of IL-1[Formula: see text] and IL-18. SA-[Formula: see text]-gal staining was used to evaluate cellular senescence. Our results show that ginsenoside Rb1 effectively suppressed BLM-induced PF in mice. ARD administration to induce cellular senescence reduced NRF2, QKI, and SMAD7 expression in MRC-5 cells. By inducing NRF2 overexpression, ARD-induced cellular senescence and fibrosis in MRC-5 cells were relieved. Notably, NRF2 knockdown abolished the mitigating effects of ginsenoside Rb1 on ARD-induced cellular senescence and fibrosis in MRC-5 cells. Mechanistically, NRF2 increased SMAD7 mRNA stability through the transcriptional regulation of QKI. As expected, ginsenoside Rb1 alleviated ARD-induced senescence and fibrosis in MRC-5 cells by activating the NRF2/QKI/SMAD7 axis. Therefore, it was found that ginsenoside Rb1 mitigates cellular senescence and fibrosis during PF progression by activating the NRF2/QKI/SMAD7 axis. This study provides a potential therapeutic strategy for the treatment of PF and elucidates its mechanism of action.
Collapse
Affiliation(s)
- Qing Zheng
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Feng-Ping Lei
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Shan Hui
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Ming Tong
- Department of Infectious Diseases, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| | - Li-Hui Liang
- Department of Geriatrics, Hunan Provincial People's Hospital, (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, P. R. China
| |
Collapse
|
11
|
Wang J, Zeng X, Xue W, Jia Q, Jiang Q, Huo C, Jiao X, Zhang J, Wang Y, Tian L, Zhu Z. Transcriptomic profiling of lung fibroblasts in silicosis: Regulatory roles of Nrf2 agonists in a mouse model. Int Immunopharmacol 2024; 143:113273. [PMID: 39362014 DOI: 10.1016/j.intimp.2024.113273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Silicosis is an occupational disease caused by long-term inhalation of free silica, resulting in a significant global health burden. Its pathogenesis remains unclear, and there is no effective treatment. Proliferative and activated myofibroblasts play a key role in the development of silicosis. Traditional studies have focused on fibroblast proliferation and collagen secretion, neglecting their functional heterogeneity. With the advancement of omics research, more pathogenic fibroblast subgroups and their functions have been identified. In this study, we applied transcriptomics to analyze gene changes in primary lung fibroblasts during silicosis development using a mouse model. Our results indicate that DEGs are enriched in collagen secretion, ECM synthesis, leukocyte migration, and chemotaxis functions. Altered core genes are associated with immune cell recruitment and cell migration. Nrf2 agonists, known for anti-inflammatory and antioxidant properties, have shown potential therapeutic effects in fibrotic diseases. However, their effects on fibroblasts in silicosis are not fully understood. We used four common Nrf2 agonists to study gene expression changes in lung fibroblasts at the transcriptome level, combined with histopathological and biochemical methods, to investigate their effects on silicosis in mice. Results show that Nrf2 agonists can exert anti-silicosis fibrosis functions by downregulating genes like Fos and Egr1, involved in cell differentiation, proliferation, and inflammation. In conclusion, this study suggests that inflammation-related co-functions of fibroblasts may be a potential mechanism in silicosis pathogenesis. Targeting Nrf2 may be a promising strategy to alleviate oxidative stress and inflammation in silicosis.
Collapse
Affiliation(s)
- Jiaxin Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xinying Zeng
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Wenming Xue
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Qiyue Jia
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Qiyue Jiang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Chuanyi Huo
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xukun Jiao
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Jiaxin Zhang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
12
|
Hu R, Wu F, Zheng YQ. Ivacaftor attenuates gentamicin-induced ototoxicity through the CFTR-Nrf2-HO1/NQO1 pathway. Redox Rep 2024; 29:2332038. [PMID: 38563333 PMCID: PMC10993751 DOI: 10.1080/13510002.2024.2332038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
OBJECTIVES Gentamicin is one of the most common ototoxic drugs that can lower patients' quality of life. Oxidative stress is a key factors inducing sensory hair cell death during gentamicin administration. So far, there are no effective drugs to prevent or treat gentamicin- induced hearing loss. A recent study found cystic fibrosis transmembrane conductance regulator (CFTR) as a new target to modulate cellular oxidative balance. The objective of this study was to estimate the effect of the CFTR activator ivacaftor on gentamicin-induced ototoxicity and determine its mechanism. METHODS The hair cell count was analyzed by Myosin 7a staining. Apoptosis was analyzed by TUNEL Apoptosis Kit. Cellular reactive oxygen species (ROS) level was detected by DCFH-DA probes. The Nrf2 related proteins expression levels were analyzed by western blot. RESULTS An in vitro cochlear explant model showed that gentamicin caused ROS accumulation in sensory hair cells and induced apoptosis, and this effect was alleviated by pretreatment with ivacaftor. Western blotting showed that ivacaftor administration markedly increased the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO1), and NAD(P)H:quinone oxidoreductase 1 (NQO1). The protective effect of ivacaftor was abolished by the Nrf2 inhibitor ML385. DISCUSSION Our results indicate the protective role of the CFTR-Nrf2-HO1/NQO1 pathway in gentamicin-induced ototoxicity. Ivacaftor may be repositioned or repurposed towards aminoglycosides-induced hearing loss.
Collapse
Affiliation(s)
- Rui Hu
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, People’s Republic of China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Fan Wu
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Pathology and Laboratory Medicine, The Medical University of South Carolina, Charleston, SC, USA
| | - Yi-Qing Zheng
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, People’s Republic of China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
13
|
Wu X, Xu H, Zhang Z, Ma Z, Zhang L, Wang C, Lan K, Li R, Chen M. Disulfiram Alleviates MTX-Induced Pulmonary Fibrosis by Inhibiting EMT in Type 2 Alveolar Epithelial Cells. Lung 2024; 203:4. [PMID: 39601871 DOI: 10.1007/s00408-024-00764-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 10/14/2024] [Indexed: 11/29/2024]
Abstract
PURPOSE Methotrexate (MTX)-induced pulmonary fibrosis is associated with high morbidity and mortality, with limited treatment options available. This study investigates whether disulfiram (DSF) can mitigate MTX-induced pulmonary fibrosis and explores the underlying mechanisms. METHODS Eight-week-old male mice were divided into control, DSF, MTX, and MTX+DSF groups and treated for 8 weeks. Weight, food, and water intake were monitored. Post-treatment, lung tissues were analyzed using HE and Masson staining, and electron microscopy. Real-time qPCR and ELISA were employed to assess inflammatory markers such as IL-1β and TNF-α in lung tissues and serum. PCR, ELISA, and Western blot were used for fibrotic markers including Col1α1, α-SMA, and hydroxyproline. Type 2 alveolar epithelial cell line MLE12 cells were similarly grouped, followed by RNA sequencing and bioinformatics analysis to elucidate the mechanisms by which DSF exerts anti-MTX-induced pulmonary fibrosis effects. ELISA and Western blot were used to measure E-cadherin and α-SMA expression. RESULTS DSF significantly reduced MTX-induced alveolar septal thickening, pulmonary fibrosis, and inflammatory cell infiltration. It also decreased the expression of inflammatory factors IL-1β and TNF-α, as well as the expression of Col1α1, α-SMA, and others. RNA-seq revealed that DSF induces changes in multiple signaling pathways associated with pulmonary fibrosis, particularly in extracellular matrix-related genes. ELISA and Western blot showed decreased E-cadherin and increased α-SMA in the MTX group, which was partially restored with DSF treatment. CONCLUSION DSF alleviates MTX-induced pulmonary fibrosis by reducing epithelial-mesenchymal transition (EMT) in type 2 alveolar epithelial cells. Disulfiram shows potential as a therapeutic agent for MTX-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiaohui Wu
- Clinical Medical School, Xi'an Medical University, Xi'an, 710021, Shaanxi, China.
| | - Hong Xu
- Department of Pathology, State Key Laboratory of Cancer Biology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhaohua Zhang
- Pharmacy School, Xi'an Medical University, Xi'an, 710021, Shaanxi, China
| | - Ziyi Ma
- Clinical Medical School, Xi'an Medical University, Xi'an, 710021, Shaanxi, China
| | - Linyi Zhang
- Clinical Medical School, Xi'an Medical University, Xi'an, 710021, Shaanxi, China
| | - Chunyang Wang
- Clinical Medical School, Xi'an Medical University, Xi'an, 710021, Shaanxi, China
| | - Kai Lan
- Clinical Medical School, Xi'an Medical University, Xi'an, 710021, Shaanxi, China
| | - Rong Li
- Clinical Medical School, Xi'an Medical University, Xi'an, 710021, Shaanxi, China
| | - Min Chen
- Clinical Medical School, Xi'an Medical University, Xi'an, 710021, Shaanxi, China
| |
Collapse
|
14
|
Niayesh-Mehr R, Kalantar M, Bontempi G, Montaldo C, Ebrahimi S, Allameh A, Babaei G, Seif F, Strippoli R. The role of epithelial-mesenchymal transition in pulmonary fibrosis: lessons from idiopathic pulmonary fibrosis and COVID-19. Cell Commun Signal 2024; 22:542. [PMID: 39538298 PMCID: PMC11558984 DOI: 10.1186/s12964-024-01925-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Despite the tremendous advancements in the knowledge of the pathophysiology and clinical aspects of SARS-CoV-2 infection, still many issues remain unanswered, especially in the long-term effects. Mounting evidence suggests that pulmonary fibrosis (PF) is one of the most severe complications associated with COVID-19. Therefore, understanding the molecular mechanisms behind its development is helpful to develop successful therapeutic strategies. Epithelial to mesenchymal transition (EMT) and its cell specific variants endothelial to mesenchymal transition (EndMT) and mesothelial to mesenchymal transition (MMT) are physio-pathologic cellular reprogramming processes induced by several infectious, inflammatory and biomechanical stimuli. Cells undergoing EMT acquire invasive, profibrogenic and proinflammatory activities by secreting several extracellular mediators. Their activity has been implicated in the pathogenesis of PF in a variety of lung disorders, including idiopathic pulmonary fibrosis (IPF) and COVID-19. Aim of this article is to provide an updated survey of the cellular and molecular mechanisms, with emphasis on EMT-related processes, implicated in the genesis of PF in IFP and COVID-19.
Collapse
Affiliation(s)
- Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Kalantar
- Department of Occupational Health, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Giulio Bontempi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Saeedeh Ebrahimi
- Department of Medical Microbiology (Bacteriology and Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghader Babaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Faezeh Seif
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy.
| |
Collapse
|
15
|
Bian Y, Yin D, Zhang P, Hong L, Yang M. Zerumbone alleviated bleomycin-induced pulmonary fibrosis in mice via SIRT1/Nrf2 pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8979-8992. [PMID: 38874804 DOI: 10.1007/s00210-024-03170-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024]
Abstract
Pulmonary fibrosis (PF) is a persistent interstitial lung condition for which effective treatment options are currently lacking. Zerumbone (zerum), a humulane sesquiterpenoid extracted from Zingiber zerumbet Smith, has been documented in previous studies to possess various pharmacological benefits. The aim of this study was to observe and investigate the therapeutic effects and mechanisms of zerum on pulmonary fibrosis. We utilized a transforming growth factor (TGF)-β1-induced human lung fibroblast (MRC-5) activation model and a bleomycin-induced pulmonary fibrosis mouse model. Cell counting kit 8 (CCK8) and cell migration assays were performed to assess the effects of zerum on MRC-5 cells. Masson's trichrome, Hematoxylin and Eosin (HE), and Sirius Red staining were conducted for pathological evaluation of lung tissue. Western blot experiments were conducted to measure the protein expression levels of Collagen I, α-SMA, Nrf2, and SIRT1. Immunofluorescence and immunohistochemistry assays were used to detect the expression of reactive oxygen species (ROS), Nrf2, and α-SMA. ELISA was employed to measure the levels of MDA, SOD, and GSH-Px. Our findings from in vitro and in vivo studies demonstrated that zerum significantly inhibited the migration ability of TGF-β1-induced MRC-5 cells, reduced ROS production in TGF-β1-induced MRC-5 cells and pulmonary fibrosis mice, and decreased the expression of Collagen I and α-SMA proteins. Additionally, zerum activated the SIRT1/Nrf2 signaling pathway in TGF-β1-induced MRC-5 cells and pulmonary fibrosis mice. Knockdown of SIRT1 abolished the anti-fibrotic effects of zerum. These results suggest that zerum inhibits TGF-β1 and BLM-induced cell and mouse pulmonary fibrosis by activating the SIRT1/Nrf2 pathway.
Collapse
Affiliation(s)
- Yali Bian
- Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Qixia District, Nanjing City, Jiangsu Province, China
| | - Dongqi Yin
- Department of Pediatrics, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Pei Zhang
- Department of Pediatrics, Chinese People's Liberation Army Eastern Theater Command General Hospital, Nanjing, Jiangsu Province, China
| | - Lingling Hong
- Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Qixia District, Nanjing City, Jiangsu Province, China
| | - Meng Yang
- Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Qixia District, Nanjing City, Jiangsu Province, China.
| |
Collapse
|
16
|
Chen W, Gao Y, Liu Y, Luo Y, Xue X, Xiao C, Wei K. Tanshinone IIA Loaded Inhaled Polymer Nanoparticles Alleviate Established Pulmonary Fibrosis. ACS Biomater Sci Eng 2024; 10:6250-6262. [PMID: 39288315 DOI: 10.1021/acsbiomaterials.4c00532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal respiratory disease characterized by chronic, progressive scarring of the lung parenchyma, leading to an irreversible decline in lung function. Apart from supportive care, there is currently no specific treatment available to reverse the disease. Based on the fact that tanshinone IIA (TAN) had an effect on protecting against TGF-β1-induced fibrosis through the inhibition of Smad and non-Smad signal pathways to avoid myofibroblasts activation, this study reported the development of the inhalable tanshinone IIA-loaded chitosan-oligosaccharides-coated poly(lactic-co-glycolic acid) (PLGA) nanoparticles (CPN@TAN) for enhancing the pulmonary delivery of tanshinone IIA to treat pulmonary fibrosis. The CPN@TAN with a size of 206.5 nm exhibited excellent in vitro aerosol delivery characteristics, featuring a mass median aerodynamic diameter (MMAD) of 3.967 ± 0.025 μm and a fine particle fraction (FPF) of 70.516 ± 0.929%. Moreover, the nanoparticles showed good stability during atomization and enhanced the mucosal penetration capabilities. The results of confocal spectroscopy confirmed the potential of the nanoparticles as carriers that facilitated the uptake of drugs by NIH3T3, A549, and MH-S cells. Additionally, the nanoparticles demonstrated good in vitro biocompatibility. In a mouse model of bleomycin-induced pulmonary fibrosis, noninvasive inhalation of aerosol CPN@TAN greatly suppressed collagen formation and facilitated re-epithelialization of the destroyed alveolar epithelium without causing systemic toxicity compared with intravenous administration. Consequently, our noninvasive inhalation drug delivery technology based on polymers may represent a promising paradigm and open the door to overcoming the difficulties associated with managing pulmonary fibrosis.
Collapse
Affiliation(s)
- Wenyu Chen
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanyuan Gao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanqi Liu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yujia Luo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xinrui Xue
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Chujie Xiao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Kun Wei
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
17
|
Lian S, Su J, Fatima I, Zhang Y, Kuang T, Hu H, Qu D, Si H, Sun W. Revealing the exceptional antioxidant activity of phosphorylated polysaccharides from medicinal Abrus cantoniensis Hance. Int J Biol Macromol 2024; 278:134532. [PMID: 39142474 DOI: 10.1016/j.ijbiomac.2024.134532] [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: 04/02/2024] [Revised: 08/04/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024]
Abstract
Abrus cantoniensis Polysaccharides (ACP) exhibit antioxidant activity and immune-regulatory functions. Abrus cantoniensis Hance widely distributed in the Guangdong and Guangxi regions of China. In this study, this research investigated the impact of phosphorylation modification on the biological activity of ACP, aiming to provide theoretical insights for its development. This research modified ACP through phosphorylation and evaluated changes in its in vitro antioxidant capacity, including free radical scavenging and resistance to cellular oxidative damage. Additionally, this research administered both native ACP and phosphorylated ACP (P-ACP) to mice to assess their protective effects against acute ethanol-induced oxidative injury. This research explored whether these effects were mediated through the Keap1-Nrf2 signaling pathway and their influence on gut microbiota. Results revealed that phosphorylation significantly enhanced ACP's antioxidant capacity and protective effects (p < 0.05). P-ACP improved mice resistance to acute oxidative injury, mitigating the adverse effects of 50 % ethanol (p < 0.05). Moreover, both ACP and P-ACP are involved in modulating the expression of the Keap1-Nrf2 signaling pathway and, to some extent, alter the composition of the gut microbiota in mice. In summary, phosphorylation modification effectively enhances ACP's antioxidant capacity and provides better protection against acute oxidative injury in mice.
Collapse
Affiliation(s)
- Shuaitao Lian
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi Grass Station, Guangxi University, Nanning 530004, Guangxi, China
| | - Jie Su
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Israr Fatima
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuan Zhang
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi Grass Station, Guangxi University, Nanning 530004, Guangxi, China
| | - Tiantian Kuang
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi Grass Station, Guangxi University, Nanning 530004, Guangxi, China
| | - Hongjie Hu
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi Grass Station, Guangxi University, Nanning 530004, Guangxi, China
| | - Dongshuai Qu
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi Grass Station, Guangxi University, Nanning 530004, Guangxi, China
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi Grass Station, Guangxi University, Nanning 530004, Guangxi, China.
| | - Wenjing Sun
- Guangxi Key Laboratory of Agricultural Resources Chemistry and Biotechnology, College of Biology & Pharmacy, Yulin Normal University, No. 1303 Jiaoyu East Road, Yulin 537000, Guangxi, China.
| |
Collapse
|
18
|
Xu W, Huang M, Dong R, Yan S, An Y, Liu B, Ma Z, Mu K, Yang Q. Anti-carbamylated protein antibodies drive AEC II toward a profibrotic phenotype by interacting with carbamylated TLR5. Rheumatology (Oxford) 2024; 63:2874-2886. [PMID: 38366924 DOI: 10.1093/rheumatology/keae111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/28/2023] [Accepted: 01/10/2024] [Indexed: 02/19/2024] Open
Abstract
OBJECTIVES This study looked at the role of anti-carbamylated protein (anti-CarP) antibodies in contributing to lung fibrosis in CTD-associated interstitial lung disease (ILD) in an autoantigen-dependent manner. METHODS ELISA was used to test serum samples, including 89 from the CTD-ILD group and 170 from the non-CTD-ILD group, for anti-CarP levels. Male C57BL/6 mice were used for the pulmonary fibrosis model and anti-CarP treatment in vivo (n = 5) and patient serum-derived or commercialized anti-CarP was used for cell treatment. We identified the carbamylated membrane protein via immunofluorescence (IF) and co-immunoprecipitation followed by mass spectrometry (MS) analysis. Quantitative RT-PCR, IF and western blot were performed to explore the antigen-dependent role of anti-CarP. A native electrophoretic mobility shift assay and MS analysis were used to verify direct interaction and carbamylation sites. RESULTS A significantly higher serum anti-CarP level was observed in CTD with ILD than without ILD. In vivo, intrapulmonary delivery of anti-CarP induces epithelial-mesenchymal transition (EMT) and microfibrotic foci. Carbamylation was enriched in type II alveolar epithelial cells (AEC II). A novel carbamylated membrane receptor, specifically recognized by anti-CarP, was identified as toll-like receptor 5 (TLR5). We found anti-CarP induces the nuclear translocation of NF-κB and downstream events, including EMT and expression of inflammatory cytokines in AEC II, which were reversed by TLR5 blocking or TLR5 knockdown. Moreover, up to 12 lysine carbamylation sites were found in TLR5 ectodomain, allowing the interaction of anti-CarP with carbamylated TLR5. CONCLUSIONS Overall, we found anti-CarP drives aberrant AEC II activation by interacting with carbamylated TLR5 to promote ILD progression.
Collapse
Affiliation(s)
- Wei Xu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Minghua Huang
- Department of Respiratory Medicine, Shandong Provincial Third Hospital, Shandong University, Jinan, China
| | - Rongrong Dong
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Suyan Yan
- Department of Rheumatology and Immunology, Shandong Province Hospital of Shandong First Medical University, Jinan, China
| | - Yan An
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baocheng Liu
- Department of Rheumatology and Immunology, Shandong Province Hospital of Shandong First Medical University, Jinan, China
| | - Zhenzhen Ma
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Rheumatology and Immunology, Shandong Province Hospital of Shandong First Medical University, Jinan, China
- Department of Integrated traditional Chinese and Western Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kun Mu
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China
| | - Qingrui Yang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Rheumatology and Immunology, Shandong Province Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
19
|
Yu H, Lou J, Ni L, Yan M, Zhu K, Mao S, Zhu J. Isoquercetin Ameliorates Osteoarthritis via Nrf2/NF-κB Axis: An In Vitro and In Vivo Study. Chem Biol Drug Des 2024; 104:e14620. [PMID: 39251394 DOI: 10.1111/cbdd.14620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/20/2024] [Accepted: 07/09/2024] [Indexed: 09/11/2024]
Abstract
Osteoarthritis (OA) is a progressive joint disease characterized by extracellular matrix (ECM) degradation and inflammation, which is involved with pathological microenvironmental alterations induced by damaged chondrocytes. However, current therapies are not effective in alleviating the progression of OA. Isoquercetin is a natural flavonoid glycoside compound that has various pharmacological effects including anticancer, anti-diabetes and blood lipid regulation. Previous evidence suggests that isoquercetin has anti-inflammatory properties in various diseases, but its effect on OA has not been investigated yet. In this study, through western bolt, qRT-PCR and ELISA, it was found that isoquercetin could reduce the increase of ADAMTS5, MMP13, COX-2, iNOS and IL-6 induced by IL-1β, suggesting that isoquercetin could inhibit the inflammation and ECM degradation of chondrocytes. Through nuclear-plasma separation technique, western blot and immunocytochemistry, it can be found that Nrf2 and NF-κB pathways are activated in this process, and isoquercetin may rely on this process to play its protective role. In vivo, the results of X-ray and SO staining show that intra-articular injection of isoquercetin reduces the degradation of cartilage in the mouse OA model. In conclusion, the present work suggests that isoquercetin may benefit chondrocytes by regulating the Nrf2/NF-κB signaling axis, which supports isoquercetin as a potential drug for the treatment of OA.
Collapse
Affiliation(s)
- He Yu
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| | - Junsheng Lou
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Libin Ni
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| | - Minwei Yan
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| | - Kewu Zhu
- Center for Drug Delivery System Research, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Su Mao
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| | - Jungao Zhu
- Department of Orthopaedics, Zhejiang Hospital, Zhejiang, Hangzhou, China
| |
Collapse
|
20
|
Li Z, Yang Y, Gao F. Monomeric compounds from natural products for the treatment of pulmonary fibrosis: a review. Inflammopharmacology 2024; 32:2203-2217. [PMID: 38724690 DOI: 10.1007/s10787-024-01485-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/23/2024] [Indexed: 08/06/2024]
Abstract
Pulmonary fibrosis (PF) is the end stage of lung injury and chronic lung diseases that results in diminished lung function, respiratory failure, and ultimately mortality. Despite extensive research, the pathogenesis of this disease remains elusive, and effective therapeutic options are currently limited, posing a significant clinical challenge. In addition, research on traditional Chinese medicine and naturopathic medicine is hampered by several complications due to complex composition and lack of reference compounds. Natural product monomers, possessing diverse biological activities and excellent safety profiles, have emerged as potential candidates for preventing and treating PF. The effective anti-PF ingredients identified can be generally divided into flavonoids, saponins, polysaccharides, and alkaloids. Specifically, these monomeric compounds can attenuate inflammatory response, oxidative stress, and other physiopathological processes of the lung through many signaling pathways. They also improve pulmonary factors. Additionally, they ameliorate epithelial-mesenchymal transition (EMT) and fibroblast-myofibroblast transdifferentiation (FMT) by regulating multiple signal amplifiers in the lungs, thereby mitigating PF. This review highlights the significant role of monomer compounds derived from natural products in reducing inflammation, oxidative stress, and inhibiting EMT process. The article provides comprehensive information and serves as a solid foundation for further exploration of new strategies to harness the potential of botanicals in the treatment of PF.
Collapse
Affiliation(s)
- Zhuqing Li
- University of Shanghai for Science and Technology, 516, Jungong Road, Shanghai, 200093, China
| | - Yanyong Yang
- Basic Medical Center for Pulmonary Disease, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, China.
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, People's Republic of China.
| | - Fu Gao
- University of Shanghai for Science and Technology, 516, Jungong Road, Shanghai, 200093, China.
- Basic Medical Center for Pulmonary Disease, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, China.
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
21
|
Kilic KD, Erisik D, Taskiran D, Turhan K, Kose T, Cetin EO, Sendemi R A, Uyanikgil Y. Protective effects of E-CG-01 (3,4-lacto cycloastragenol) against bleomycin-induced lung fibrosis in C57BL/6 mice. Biomed Pharmacother 2024; 177:117016. [PMID: 38943992 DOI: 10.1016/j.biopha.2024.117016] [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: 04/15/2024] [Revised: 06/05/2024] [Accepted: 06/17/2024] [Indexed: 07/01/2024] Open
Abstract
Idiopathic pulmonary fibrosis is an aging-related, chronic lung disease, with unclear pathogenesis and no effective treatment. One of the triggering factors in cell aging is oxidative stress and it is known to have a role in idiopathic pulmonary fibrosis. In this paper, the protective effect of the E-CG-01 (3,4-lacto-cycloastragenol) molecule in terms of its antioxidant properties was evaluated in the bleomycin induced mice lung fibrosis model. Bleomycin sulfate was administered as a single dose (2.5 U/kg body weight) intratracheally to induce lung fibrosis. E-CG-01 was administered intraperitoneally in three different doses (2 mg/kg/day, 6 mg/kg/day, and 10 mg/kg/day) for 14 days, starting three days before the bleomycin administration. Fibrosis was examined by Hematoxylin-Eosin, Masson Trichrome, and immunohistochemical staining for TGF-beta1, Type I collagen Ki-67, and gama-H2AX markers. Activity analysis of catalase and Superoxide dismutase enzymes, measurement of total oxidant, total glutathione, and Malondialdehyde levels. In histological analysis, it was determined that all three different doses of the molecule provided a prophylactic effect against the progression of fibrosis compared to the bleomycin control group. However, it was observed that only the molecule applied in the high dose decreased the total oxidant stress level. Lung weight ratio increased in the BLM group but significantly reduced with high-dose E-CG-01. E-CG-01 at all doses reduced collagen deposition, TGF-β expression, and Ki-67 expression compared to the BLM group. Intermediate and high doses of E-CG-01 also significantly reduced alveolar wall thickness and edema formation. These findings suggest that E-CG-01 has potential therapeutic effects in mitigating lung fibrosis through its antioxidant properties.
Collapse
Affiliation(s)
- Kubilay Dogan Kilic
- Ege University, Faculty of Medicine, Department of Histology and Embryology, İzmir, Turkiye; Leibniz Institute for Evolution and Biodiversity Science, Museum für Naturkunde, Berlin, Germany.
| | - Derya Erisik
- Ege University, Faculty of Medicine, Department of Histology and Embryology, İzmir, Turkiye
| | - Dilek Taskiran
- Ege University, Faculty of Medicine, Department of Physiology, İzmir, Turkiye
| | - Kutsal Turhan
- Ege University, Faculty of Medicine, Department of Thoracic Surgery, İzmir, Turkiye; Acibadem Kent Hospital, Department of Thoracic Surgery, İzmir, Türkiye
| | - Timur Kose
- Ege University, Faculty of Medicine, Department of Biostatistics and Medical Informatics, İzmir, Turkiye
| | - Emel Oyku Cetin
- Ege University, Faculty of Pharmacy, Department of Biopharmaceutics and Pharmacokinetics, İzmir, Turkiye
| | - Aylin Sendemi R
- Ege University, Faculty of Engineering, Department of Bioengineering, İzmir, Turkiye
| | - Yiğit Uyanikgil
- Ege University, Faculty of Medicine, Department of Histology and Embryology, İzmir, Turkiye; Ege University, Cord Blood Cell - Tissue Research and Application Center, İzmir, Turkiye; Ege University, Institute of Health Sciences, Department of Stem Cell, İzmir, Turkiye
| |
Collapse
|
22
|
林 雅, 颜 崇, 洪 文, 蔡 成, 龚 小. [Role and mechanism of epithelial-mesenchymal transition in a rat model of bronchopulmonary dysplasia induced by hyperoxia exposure]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:765-773. [PMID: 39014955 PMCID: PMC11562037 DOI: 10.7499/j.issn.1008-8830.2312112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/31/2024] [Indexed: 07/18/2024]
Abstract
OBJECTIVES To investigate the role and mechanism of epithelial-mesenchymal transition (EMT) in a rat model of bronchopulmonary dysplasia (BPD). METHODS The experiment consisted of two parts. (1) Forty-eight preterm rats were randomly divided into a normoxia group and a hyperoxia group, with 24 rats in each group. The hyperoxia group was exposed to 85% oxygen to establish a BPD model, while the normoxia group was kept in room air at normal pressure. Lung tissue samples were collected on days 1, 4, 7, and 14 of the experiment. (2) Rat type II alveolar epithelial cells (RLE-6TN) were randomly divided into a normoxia group (cultured in air) and a hyperoxia group (cultured in 95% oxygen), and cell samples were collected 12, 24, and 48 hours after hyperoxia exposure. Hematoxylin-eosin staining was used to observe alveolarization in preterm rat lungs, and immunofluorescence was used to detect the co-localization of surfactant protein C (SPC) and α-smooth muscle actin (α-SMA) in preterm rat lung tissue and RLE-6TN cells. Quantitative real-time polymerase chain reaction and protein immunoblotting were used to detect the expression levels of EMT-related mRNA and proteins in preterm rat lung tissue and RLE-6TN cells. RESULTS (1) Compared with the normoxia group, the hyperoxia group showed blocked alveolarization and simplified alveolar structure after 7 days of hyperoxia exposure. Co-localization of SPC and α-SMA was observed in lung tissue, with decreased SPC expression and increased α-SMA expression in the hyperoxia group at 7 and 14 days of hyperoxia exposure compared to the normoxia group. In the hyperoxia group, the mRNA and protein levels of TGF-β1, α-SMA, and N-cadherin were increased, while the mRNA and protein levels of SPC and E-cadherin were decreased at 7 and 14 days of hyperoxia exposure compared to the normoxia group (P<0.05). (2) SPC and α-SMA was observed in RLE-6TN cells, with decreased SPC expression and increased α-SMA expression in the hyperoxia group at 24 and 48 hours of hyperoxia exposure compared to the normoxia group. Compared to the normoxia group, the mRNA and protein levels of SPC and E-cadherin in the hyperoxia group were decreased, while the mRNA and protein levels of TGF-β1, α-SMA, and E-cadherin in the hyperoxia group increased at 48 hours of hyperoxia exposure (P<0.05). CONCLUSIONS EMT disrupts the tight connections between alveolar epithelial cells in a preterm rat model of BPD, leading to simplified alveolar structure and abnormal development, and is involved in the development of BPD. Citation:Chinese Journal of Contemporary Pediatrics, 2024, 26(7): 765-773.
Collapse
|
23
|
Shen S, Hu M, Peng Y, Zheng Y, Zhang R. Research Progress in pathogenesis of connective tissue disease-associated interstitial lung disease from the perspective of pulmonary cells. Autoimmun Rev 2024; 23:103600. [PMID: 39151642 DOI: 10.1016/j.autrev.2024.103600] [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: 04/14/2024] [Revised: 07/16/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024]
Abstract
The lungs are a principal factor in the increased morbidity and mortality observed in patients with Connective Tissue Disease (CTD), frequently presenting as CTD-associated Interstitial Lung Disease (ILD). Currently, there is a lack of comprehensive descriptions of the pulmonary cells implicated in the development of CTD-ILD. This review leverages the Human Lung Cell Atlas (HLCA) and spatial multi-omics atlases to discuss the advancements in research on the pathogenesis of CTD-ILD from a pulmonary cell perspective. This facilitates a more precise localization of disease sites and a more systematic consideration of disease progression, supporting further mechanistic studies and targeted therapies.
Collapse
Affiliation(s)
- Shuyi Shen
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Ming Hu
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yi Peng
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yi Zheng
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Rong Zhang
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| |
Collapse
|
24
|
Cao W, Chen Z, Lin C, Lin X, Chen Y, Zhang J. Honokiol Mitigates Metabolic-Associated Fatty Liver Disease by Regulating Nrf2 and RIPK3 Signaling Pathways. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35:551-559. [PMID: 39128113 PMCID: PMC11363389 DOI: 10.5152/tjg.2024.23470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/13/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND/AIMS Metabolic-associated fatty liver disease (MAFLD) is a common cause of chronic liver disease worldwide. However, there is currently no recognized effective drugs for treating it. MATERIALS AND METHODS In this study, we investigated the efficacy of Honokiol (HNK) in vitro for mitigating MAFLD. Then, 0.4 mM palmitic acid (PA) and LO2 cells were used to establish the MAFLD model. The protective effect of HNK on MAFLD was confirmed by Oil Red O staining and cell counting kit (CCK-8) assay in LO2 cell line. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were carried out to analyze the regulatory role of HNK on Nrf2 and RIPK3 signaling pathways. The effect of HNK and its downstream signaling pathways on oxidative stress were verified by the detection of reactive oxygen species (ROS), malondialdehyde (MDA), catalase (CAT), and superoxide dismutase (SOD). The concentration of IL-1β, IL-6L, and TNF-α was assessed by enzyme-linked immunosorbent assay (ELISA). RESULTS The middle concentration of HNK (50 μmol/L) was selected as the best option for inhibiting lipidosis and oxidative stress in MAFLD models. Honokiol mitigates MAFLD via activation of nuclear factor E2-related factor 2 (Nrf2) signaling pathways in vitro. Honokiol suppressed MAFLD via activating the Nrf2 signaling pathway to play an antioxidant and anti-inflammatory role. Also, HNK regulates Nrf2 and RIPK3 signaling pathways to mitigate MAFLD. CONCLUSION Our results showed that HNK may suppress the oxidative stress and inflammation in MAFLD via activation of Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Wen Cao
- Department of Gastroenterology, Fuzhou Second General Hospital, Fuzhou, Fujian Province, China
| | - Zengdian Chen
- Department of Gastroenterology, Fuzhou Second General Hospital, Fuzhou, Fujian Province, China
| | - Chenhui Lin
- Department of Gastroenterology, Fuzhou Second General Hospital, Fuzhou, Fujian Province, China
| | - Xiaojin Lin
- Department of Gastroenterology, Fuzhou Second General Hospital, Fuzhou, Fujian Province, China
| | - Yang Chen
- Department of Gastroenterology, Fuzhou Second General Hospital, Fuzhou, Fujian Province, China
| | - Jingjuan Zhang
- Department of Gastroenterology, Fuzhou Second General Hospital, Fuzhou, Fujian Province, China
| |
Collapse
|
25
|
Gao Y, Liu MF, Li Y, Liu X, Cao YJ, Long QF, Yu J, Li JY. Mesenchymal stem cells-extracellular vesicles alleviate pulmonary fibrosis by regulating immunomodulators. World J Stem Cells 2024; 16:670-689. [PMID: 38948098 PMCID: PMC11212550 DOI: 10.4252/wjsc.v16.i6.670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/22/2024] [Accepted: 05/11/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a chronic interstitial lung disease characterized by fibroblast proliferation and extracellular matrix formation, causing structural damage and lung failure. Stem cell therapy and mesenchymal stem cells-extracellular vesicles (MSC-EVs) offer new hope for PF treatment. AIM To investigate the therapeutic potential of MSC-EVs in alleviating fibrosis, oxidative stress, and immune inflammation in A549 cells and bleomycin (BLM)-induced mouse model. METHODS The effect of MSC-EVs on A549 cells was assessed by fibrosis markers [collagen I and α-smooth muscle actin (α-SMA), oxidative stress regulators [nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), and inflammatory regulators [nuclear factor-kappaB (NF-κB) p65, interleukin (IL)-1β, and IL-2]. Similarly, they were assessed in the lungs of mice where PF was induced by BLM after MSC-EV transfection. MSC-EVs ion PF mice were detected by pathological staining and western blot. Single-cell RNA sequencing was performed to investigate the effects of the MSC-EVs on gene expression profiles of macrophages after modeling in mice. RESULTS Transforming growth factor (TGF)-β1 enhanced fibrosis in A549 cells, significantly increasing collagen I and α-SMA levels. Notably, treatment with MSC-EVs demonstrated a remarkable alleviation of these effects. Similarly, the expression of oxidative stress regulators, such as Nrf2 and HO-1, along with inflammatory regulators, including NF-κB p65 and IL-1β, were mitigated by MSC-EV treatment. Furthermore, in a parallel manner, MSC-EVs exhibited a downregulatory impact on collagen deposition, oxidative stress injuries, and inflammatory-related cytokines in the lungs of mice with PF. Additionally, the mRNA sequencing results suggested that BLM may induce PF in mice by upregulating pulmonary collagen fiber deposition and triggering an immune inflammatory response. The findings collectively highlight the potential therapeutic efficacy of MSC-EVs in ameliorating fibrotic processes, oxidative stress, and inflammatory responses associated with PF. CONCLUSION MSC-EVs could ameliorate fibrosis in vitro and in vivo by downregulating collagen deposition, oxidative stress, and immune-inflammatory responses.
Collapse
Affiliation(s)
- Ying Gao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial Rehabilitation Hospital, Xi'an 710000, Shaanxi Province, China
| | - Mei-Fang Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Ningxia Medical University (The First People's Hospital of Yinchuan), Yinchuan 750001, Ningxia Hui Autonomous Region, China
| | - Yang Li
- School of Clinical Medicine, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
| | - Xi Liu
- Department of Respiratory and Critical Care Medicine, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Yu-Jie Cao
- Department of Respiratory and Critical Care Medicine, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Qian-Fa Long
- Department of Neurosurgery, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Jun Yu
- Department of Emergency, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Jian-Ying Li
- Department of Respiratory and Critical Care Medicine, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China.
| |
Collapse
|
26
|
Gao Y, Liu MF, Li Y, Liu X, Cao YJ, Long QF, Yu J, Li JY. Mesenchymal stem cells-extracellular vesicles alleviate pulmonary fibrosis by regulating immunomodulators. World J Stem Cells 2024; 16:669-688. [DOI: 10.4252/wjsc.v16.i6.669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/22/2024] [Accepted: 05/11/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a chronic interstitial lung disease characterized by fibroblast proliferation and extracellular matrix formation, causing structural damage and lung failure. Stem cell therapy and mesenchymal stem cells-extracellular vesicles (MSC-EVs) offer new hope for PF treatment.
AIM To investigate the therapeutic potential of MSC-EVs in alleviating fibrosis, oxidative stress, and immune inflammation in A549 cells and bleomycin (BLM)-induced mouse model.
METHODS The effect of MSC-EVs on A549 cells was assessed by fibrosis markers [collagen I and α-smooth muscle actin (α-SMA), oxidative stress regulators [nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), and inflammatory regulators [nuclear factor-kappaB (NF-κB) p65, interleukin (IL)-1β, and IL-2]. Similarly, they were assessed in the lungs of mice where PF was induced by BLM after MSC-EV transfection. MSC-EVs ion PF mice were detected by pathological staining and western blot. Single-cell RNA sequencing was performed to investigate the effects of the MSC-EVs on gene expression profiles of macrophages after modeling in mice.
RESULTS Transforming growth factor (TGF)-β1 enhanced fibrosis in A549 cells, significantly increasing collagen I and α-SMA levels. Notably, treatment with MSC-EVs demonstrated a remarkable alleviation of these effects. Similarly, the expression of oxidative stress regulators, such as Nrf2 and HO-1, along with inflammatory regulators, including NF-κB p65 and IL-1β, were mitigated by MSC-EV treatment. Furthermore, in a parallel manner, MSC-EVs exhibited a downregulatory impact on collagen deposition, oxidative stress injuries, and inflammatory-related cytokines in the lungs of mice with PF. Additionally, the mRNA sequencing results suggested that BLM may induce PF in mice by upregulating pulmonary collagen fiber deposition and triggering an immune inflammatory response. The findings collectively highlight the potential therapeutic efficacy of MSC-EVs in ameliorating fibrotic processes, oxidative stress, and inflammatory responses associated with PF.
CONCLUSION MSC-EVs could ameliorate fibrosis in vitro and in vivo by downregulating collagen deposition, oxidative stress, and immune-inflammatory responses.
Collapse
Affiliation(s)
- Ying Gao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial Rehabilitation Hospital, Xi’an 710000, Shaanxi Province, China
| | - Mei-Fang Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Ningxia Medical University (The First People’s Hospital of Yinchuan), Yinchuan 750001, Ningxia Hui Autonomous Region, China
| | - Yang Li
- School of Clinical Medicine, Xi’an Medical University, Xi’an 710021, Shaanxi Province, China
| | - Xi Liu
- Department of Respiratory and Critical Care Medicine, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| | - Yu-Jie Cao
- Department of Respiratory and Critical Care Medicine, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| | - Qian-Fa Long
- Department of Neurosurgery, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| | - Jun Yu
- Department of Emergency, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| | - Jian-Ying Li
- Department of Respiratory and Critical Care Medicine, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| |
Collapse
|
27
|
Wang Y, He X, Cheng N, Huang K. Unveiling the Nutritional Veil of Sulforaphane: With a Major Focus on Glucose Homeostasis Modulation. Nutrients 2024; 16:1877. [PMID: 38931232 PMCID: PMC11206418 DOI: 10.3390/nu16121877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Abnormal glucose homeostasis is associated with metabolic syndromes including cardiovascular diseases, hypertension, type 2 diabetes mellitus, and obesity, highlighting the significance of maintaining a balanced glucose level for optimal biological function. This highlights the importance of maintaining normal glucose levels for proper biological functioning. Sulforaphane (SFN), the primary bioactive compound in broccoli from the Cruciferae or Brassicaceae family, has been shown to enhance glucose homeostasis effectively while exhibiting low cytotoxicity. This paper assesses the impact of SFN on glucose homeostasis in vitro, in vivo, and human trials, as well as the molecular mechanisms that drive its regulatory effects. New strategies have been proposed to enhance the bioavailability and targeted delivery of SFN in order to overcome inherent instability. The manuscript also covers the safety evaluations of SFN that have been documented for its production and utilization. Hence, a deeper understanding of the favorable influence and mechanism of SFN on glucose homeostasis, coupled with the fact that SFN is abundant in the human daily diet, may ultimately offer theoretical evidence to support its potential use in the food and pharmaceutical industries.
Collapse
Affiliation(s)
- Yanan Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.W.); (X.H.); (N.C.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.W.); (X.H.); (N.C.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| | - Nan Cheng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.W.); (X.H.); (N.C.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.W.); (X.H.); (N.C.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| |
Collapse
|
28
|
Shen W, Li C, Liu Q, Cai J, Wang Z, Pang Y, Ning G, Yao X, Kong X, Feng S. Celastrol inhibits oligodendrocyte and neuron ferroptosis to promote spinal cord injury recovery. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155380. [PMID: 38507854 DOI: 10.1016/j.phymed.2024.155380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/01/2024] [Accepted: 01/17/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Spinal cord injury (SCI) is a traumatic injury to the central nervous system and can cause lipid peroxidation in the spinal cord. Ferroptosis, an iron-dependent programmed cell death, plays a key role in the pathophysiology progression of SCI. Celastrol, a widely used antioxidant drug, has potential therapeutic value for nervous system. PURPOSE To investigate whether celastrol can be a reliable candidate for ferroptosis inhibitor and the molecular mechanism of celastrol in repairing SCI by inhibiting ferroptosis. METHODS First, a rat SCI model was constructed, and the recovery of motor function was observed after treatment with celastrol. The regulatory effect of celastrol on ferroptosis pathway Nrf2-xCT-GPX4 was detected by Western blot and immunofluorescence. Finally, the ferroptosis model of neurons and oligodendrocytes was constructed in vitro to further verify the mechanism of inhibiting ferroptosis by celastrol. RESULTS Our results demonstrated that celastrol promoted the recovery of spinal cord tissue and motor function in SCI rats. Further in vitro and in vivo studies showed that celastrol significantly inhibited ferroptosis in neurons and oligodendrocytes and reduced the accumulation of ROS. Finally, we found that celastrol could inhibit ferroptosis by up-regulating the Nrf2-xCT-GPX4 axis to repair SCI. CONCLUSION Celastrol effectively inhibits ferroptosis after SCI by upregulating the Nrf2-xCT-GPX4 axis, reducing the production of lipid ROS, protecting the survival of neurons and oligodendrocytes, and improving the functional recovery.
Collapse
Affiliation(s)
- Wenyuan Shen
- Spine Surgery Department of the Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, PR China; Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, PR China; Orthopedic Research Center of Shandong University & Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250063, PR China
| | - Chuanhao Li
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Quan Liu
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Jun Cai
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, 300020, PR China
| | - Zhishuo Wang
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Yilin Pang
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Guangzhi Ning
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Xue Yao
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, PR China; Orthopedic Research Center of Shandong University & Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250063, PR China.
| | - Xiaohong Kong
- Orthopedic Research Center of Shandong University & Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250063, PR China.
| | - Shiqing Feng
- Spine Surgery Department of the Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, PR China; Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, PR China; Orthopedic Research Center of Shandong University & Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250063, PR China.
| |
Collapse
|
29
|
Yang W, Liang Y, Liu Y, Yao Y, Yu Z, Chen B, Cai Y, Wei M, Zheng G. Enhancement of hepatoprotective activity of limonin from citrus seeds against acetaminophen-induced liver injury by HSCCC purification and liposomal encapsulation. Fitoterapia 2024; 175:105899. [PMID: 38471575 DOI: 10.1016/j.fitote.2024.105899] [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: 10/18/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Limonin is a natural tetracyclic triterpenoid compound in citrus seeds that presents hepatoprotective effects but is often discarded as agricultural waste because of its low content and low solubility. Herein, limonin with high purity (98.11%) from citrus seeds was obtained via purification by high-speed counter-current chromatography (HSCCC) and recrystallization. Limonin-loaded liposomes (Lip-LM) prepared by thin film hydration and high pressure homogenization method to enhance its solubility and hepatoprotective effect on APAP-induced liver injury (AILI). Lip-LM appeared as lipid nanoparticles under a transmission electron microscope, and showed well dispersed nano-scale size (69.04 ± 0.42 nm), high encapsulation efficiency (93.67% ± 2.51%), sustained release, fine stability. Lip-LM also exhibited significantly better hepatoprotective activity on AILI than free limonin in vivo. In summary, Lip-LM might be used as a potential hepatoprotective agent in the form of dietary supplement and provide an effective strategy to improve the potential value of citrus seeds.
Collapse
Affiliation(s)
- Wanling Yang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510645, China
| | - Yiyao Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510645, China
| | - Yujie Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510645, China
| | - Yunan Yao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510645, China
| | - Zhiqian Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510645, China
| | - Baizhong Chen
- Guangdong Xinbaotang Biological Technology Co., Ltd, Guangdong, Jiangmen 529000, China
| | - Yi Cai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510645, China.
| | - Minyan Wei
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510645, China.
| | - Guodong Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510645, China.
| |
Collapse
|
30
|
Cai L, Wang J, Yi X, Yu S, Wang C, Zhang L, Zhang X, Cheng L, Ruan W, Dong F, Su P, Shi Y. Nintedanib-loaded exosomes from adipose-derived stem cells inhibit pulmonary fibrosis induced by bleomycin. Pediatr Res 2024; 95:1543-1552. [PMID: 38245633 DOI: 10.1038/s41390-024-03024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/05/2023] [Accepted: 12/26/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a progressive lung disorder with a high mortality rate; its therapy remains limited due to the inefficiency of drug delivery. In this study, the system of drug delivery of nintedanib (Nin) by exosomes derived from adipose-derived stem cells (ADSCs-Exo, Exo) was developed to effectively deliver Nin to lung lesion tissue to ensure enhanced anti-fibrosis therapy. METHODS The bleomycin (BLM)-induced PF model was constructed in vivo and in vitro. The effects of Exo-Nin on BLM-induced PF and its regulatory mechanism were examined using RT-qPCR, Western blotting, immunofluorescence, and H&E staining. RESULTS We found Exo-Nin significantly improved BLM-induced PF in vivo and in vitro compared to Nin and Exo groups alone. Mechanistically, Exo-Nin alleviated fibrogenesis by suppressing endothelial-mesenchymal transition through the down-regulation of the TGF-β/Smad pathway and the attenuation of oxidative stress in vivo and in vitro. CONCLUSIONS Utilizing adipose stem cell-derived exosomes as carriers for Nin exhibited a notable enhancement in therapeutic efficacy. This improvement can be attributed to the regenerative properties of exosomes, indicating promising prospects for adipose-derived exosomes in cell-free therapies for PF. IMPACT The system of drug delivery of nintedanib (Nin) by exosomes derived from adipose-derived stem cells was developed to effectively deliver Nin to lung lesion tissue to ensure enhanced anti-fibrosis therapy. The use of adipose stem cell-derived exosomes as the carrier of Nin may increase the therapeutic effect of Nin, which can be due to the regenerative properties of the exosomes and indicate promising prospects for adipose-derived exosomes in cell-free therapies for PF.
Collapse
Affiliation(s)
- Liyun Cai
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Jie Wang
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Xue Yi
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Shuwei Yu
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Chong Wang
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Liyuan Zhang
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Xiaoling Zhang
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Lixian Cheng
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Wenwen Ruan
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Feige Dong
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Ping Su
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China
| | - Ying Shi
- Department of Basic Medicine, Institute of Respiratory Diseases, Xiamen Medical College, Xiamen, China.
| |
Collapse
|
31
|
Chen YY, Wang M, Zuo CY, Mao MX, Peng XC, Cai J. Nrf-2 as a novel target in radiation induced lung injury. Heliyon 2024; 10:e29492. [PMID: 38665580 PMCID: PMC11043957 DOI: 10.1016/j.heliyon.2024.e29492] [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: 10/16/2023] [Revised: 03/09/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Radiation-induced lung injury (RILI) is a common and fatal complication of chest radiotherapy. The underlying mechanisms include radiation-induced oxidative stress caused by damage to the deoxyribonucleic acid (DNA) and production of reactive oxygen species (ROS), resulting in apoptosis of lung and endothelial cells and recruitment of inflammatory cells and myofibroblasts expressing NADPH oxidase to the site of injury, which in turn contribute to oxidative stress and cytokine production. Nuclear factor erythroid 2-related factor 2 (Nrf-2) is a vital transcription factor that regulates oxidative stress and inhibits inflammation. Studies have shown that Nrf-2 protects against radiation-induced lung inflammation and fibrosis. This review discusses the protective role of Nrf-2 in RILI and its possible mechanisms.
Collapse
Affiliation(s)
- Yuan-Yuan Chen
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Meng Wang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Chen-Yang Zuo
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Meng-Xia Mao
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, PR China
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Jun Cai
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| |
Collapse
|
32
|
Zeng Q, Wen BB, Liu X, Luo YY, Hu ZG, Huang L, Zhang XH, Huang XT, Zhou TT, Sang XX, Luo YY, Xiong DY, Luo ZQ, Liu W, Tang SY. NBR1-p62-Nrf2 mediates the anti-pulmonary fibrosis effects of protodioscin. Chin Med 2024; 19:60. [PMID: 38589903 PMCID: PMC11003024 DOI: 10.1186/s13020-024-00930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/31/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis is a persistent disease of the lung interstitium for which there is no efficacious pharmacological therapy. Protodioscin, a steroidal saponin, possesses diverse pharmacological properties; however, its function in pulmonary fibrosis is yet to be established. Hence, in this investigation, it was attempted to figure out the anti-pulmonary fibrosis influences of protodioscin and its pharmacological properties related to oxidative stress. METHODS A mouse lung fibrosis model was generated using tracheal injections of bleomycin, followed by intraperitoneal injection of different concentrations of protodioscin, and the levels of oxidative stress and fibrosis were detected in the lungs. Multiple fibroblasts were treated with TGF-β to induce their transition to myofibroblasts. It was attempted to quantify myofibroblast markers' expression levels and reactive oxygen species levels as well as Nrf2 activation after co-incubation of TGF-β with fibroblasts and different concentrations of protodioscin. The influence of protodioscin on the expression and phosphorylation of p62, which is associated with Nrf2 activation, were detected, and p62 related genes were predicted by STRING database. The effects of Nrf2 inhibitor or silencing of the Nrf2, p62 and NBR1 genes, respectively, on the activation of Nrf2 by protodioscin were examined. The associations between p62, NBR1, and Keap1 in the activation of Nrf2 by protodioscin was demonstrated using a co-IP assay. Nrf2 inhibitor were used when protodioscin was treated in mice with pulmonary fibrosis and lung tissue fibrosis and oxidative stress levels were detected. RESULTS In vivo, protodioscin decreased the levels of fibrosis markers and oxidative stress markers and activated Nrf2 in mice with pulmonary fibrosis, and these effects were inhibited by Nrf2 inhibitor. In vitro, protodioscin decreased the levels of myofibroblast markers and oxidative stress markers during myofibroblast transition and promoted Nrf2 downstream gene expression, with reversal of these effects after Nrf2, p62 and NBR1 genes were silenced or Nrf2 inhibitors were used, respectively. Protodioscin promoted the binding of NBR1 to p62 and Keap1, thereby reducing Keap1-Nrf2 binding. CONCLUSION The NBR1-p62-Nrf2 axis is targeted by protodioscin to reduce oxidative stress and inhibit pulmonary fibrosis.
Collapse
Affiliation(s)
- Qian Zeng
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Bin-Bin Wen
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xin Liu
- The Orthopedics Hospital of Traditional Chinese Medicine Zhuzhou City, Zhuzhou, Hunan, China
| | - Yong-Yu Luo
- Guiyang Second People's Hospital, Guiyang, Guizhou, China
| | - Zhen-Gang Hu
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Huang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Hua Zhang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Ting-Ting Zhou
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiao-Xue Sang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yu-Yang Luo
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Da-Yan Xiong
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Zi-Qiang Luo
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
33
|
Zheng M, Zhu W, Gao F, Zhuo Y, Zheng M, Wu G, Feng C. Novel inhalation therapy in pulmonary fibrosis: principles, applications and prospects. J Nanobiotechnology 2024; 22:136. [PMID: 38553716 PMCID: PMC10981316 DOI: 10.1186/s12951-024-02407-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
Pulmonary fibrosis (PF) threatens millions of people worldwide with its irreversible progression. Although the underlying pathogenesis of PF is not fully understood, there is evidence to suggest that the disease can be blocked at various stages. Inhalation therapy has been applied for lung diseases such as asthma and chronic obstructive pulmonary disease, and its application for treating PF is currently under consideration. New techniques in inhalation therapy, such as the application of microparticles and nanoparticles, traditional Chinese medicine monomers, gene therapy, inhibitors, or agonists of signaling pathways, extracellular vesicle interventions, and other specific drugs, are effective in treating PF. However, the safety and effectiveness of these therapeutic techniques are influenced by the properties of inhaled particles, biological and pathological barriers, and the type of inhalation device used. This review provides a comprehensive overview of the pharmacological, pharmaceutical, technical, preclinical, and clinical experimental aspects of novel inhalation therapy for treating PF and focus on therapeutic methods that significantly improve existing technologies or expand the range of drugs that can be administered via inhalation. Although inhalation therapy for PF has some limitations, the advantages are significant, and further research and innovation about new inhalation techniques and drugs are encouraged.
Collapse
Affiliation(s)
- Meiling Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100010, China
- Peking University People's Hospital, Beijing, 100032, China
| | - Wei Zhu
- Department of Ophthalmology, Changshu No. 2 People's Hospital, Changshu, 215500, China
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yu Zhuo
- Department of Medical Oncology Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100010, China
| | - Mo Zheng
- Department of Medical Oncology Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100010, China
| | - Guanghao Wu
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China.
| | - Cuiling Feng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100010, China.
- Peking University People's Hospital, Beijing, 100032, China.
| |
Collapse
|
34
|
Xie W, Deng L, Qian R, Huang X, Liu W, Tang S. Curculigoside Attenuates Endoplasmic Reticulum Stress-Induced Epithelial Cell and Fibroblast Senescence by Regulating the SIRT1-P300 Signaling Pathway. Antioxidants (Basel) 2024; 13:420. [PMID: 38671868 PMCID: PMC11047561 DOI: 10.3390/antiox13040420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
The senescence of alveolar epithelial cells (AECs) and fibroblasts plays a pivotal role in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a condition lacking specific therapeutic interventions. Curculigoside (CCG), a prominent bioactive constituent of Curculigo, exhibits anti-osteoporotic and antioxidant activities. Our investigation aimed to elucidate the anti-senescence and anti-fibrotic effects of CCG in experimental pulmonary fibrosis and delineate its underlying molecular mechanisms. Our findings demonstrate that CCG attenuates bleomycin-induced pulmonary fibrosis and lung senescence in murine models, concomitantly ameliorating lung function impairment. Immunofluorescence staining for senescence marker p21, alongside SPC or α-SMA, suggested that CCG's mitigation of lung senescence correlates closely with the deceleration of senescence in AECs and fibroblasts. In vitro, CCG mitigated H2O2-induced senescence in AECs and the natural senescence of primary mouse fibroblasts. Mechanistically, CCG can upregulate SIRT1 expression, downregulating P300 expression, enhancing Trim72 expression to facilitate P300 ubiquitination and degradation, reducing the acetylation levels of antioxidant enzymes, and upregulating their expression levels. These actions collectively inhibited endoplasmic reticulum stress (ERS) and alleviated senescence. Furthermore, the anti-senescence effects and mechanisms of CCG were validated in a D-galactose (D-gal)-induced progeroid model. This study provides novel insights into the mechanisms underlying the action of CCG in cellular senescence and chronic diseases, offering potential avenues for the development of innovative drugs or therapeutic strategies.
Collapse
Affiliation(s)
- Weixi Xie
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Lang Deng
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Rui Qian
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Xiaoting Huang
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha 410013, China; (W.X.); (L.D.); (R.Q.); (X.H.)
- The School of Nursing, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
35
|
Hui Q, Yang N, Xiong C, Zhou S, Zhou X, Jin Q, Xu X. Isorhamnetin suppresses the epithelial-mesenchymal transition of the retinal pigment epithelium both in vivo and in vitro through Nrf2-dependent AKT/GSK-3β pathway. Exp Eye Res 2024; 240:109823. [PMID: 38331017 DOI: 10.1016/j.exer.2024.109823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Age-related macular degeneration (AMD) is a major cause of blindness in the elderly worldwide. Multiple studies have shown that epithelial-mesenchymal transition (EMT) plays a pivotal role in the pathogenesis of AMD. Isorhamnetin (Isor) is a flavonoid compound that inhibits EMT in tumor cells. However, whether it can also attenuate EMT in the retinal pigment epithelium (RPE) is unknown. Therefore, our study was designed to probe the possible impact of Isor on EMT process in both mouse retina and ARPE-19 cells. C57BL/6 mice were utilized to establish a dry AMD model. Isor and LCZ (a mixture of luteine/β-carotene/zinc gluconate) were administered orally for 3 months. The effects of Isor on the retina were evaluated using fundus autofluorescence, optical coherence tomography, and transmission electron microscopy. Transwell and wound healing assay were employed to assess ARPE-19 cell migration. Western blotting and immunofluorescence were used to measure the protein expressions associated with EMT, Nrf2 and AKT/GSK-3β pathway. The findings indicated that Isor alleviated dry AMD-like pathological changes in vehicle mice retina, inhibited the migration of Ox-LDL-treated ARPE-19 cells, and repressed the EMT processes in vivo and in vitro. Furthermore, Isor activated Nrf2 pathway and deactivated AKT/GSK-3β pathway in both vehicle mice and ARPE-19 cells. Interestingly, when Nrf2 siRNA was transfected into ARPE-19 cells, the inhibitory effect of Isor on EMT and AKT/GSK-3β pathway was attenuated. These results suggested that Isor inhibited EMT processes via Nrf2-dependent AKT/GSK-3β pathway and is a promising candidate for dry AMD treatment.
Collapse
Affiliation(s)
- Qinyi Hui
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Ning Yang
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Caijian Xiong
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Siqi Zhou
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Xin Zhou
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Qingzi Jin
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China
| | - Xinrong Xu
- Department of Ophthalmology, Jiangsu Province Hospital of Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, 210029, China.
| |
Collapse
|
36
|
Azeredo PDS, Fan D, Murphy EA, Carver WE. Potential of Plant-Derived Compounds in Preventing and Reversing Organ Fibrosis and the Underlying Mechanisms. Cells 2024; 13:421. [PMID: 38474385 PMCID: PMC10930795 DOI: 10.3390/cells13050421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Increased production of extracellular matrix is a necessary response to tissue damage and stress. In a normal healing process, the increase in extracellular matrix is transient. In some instances; however, the increase in extracellular matrix can persist as fibrosis, leading to deleterious alterations in organ structure, biomechanical properties, and function. Indeed, fibrosis is now appreciated to be an important cause of mortality and morbidity. Extensive research has illustrated that fibrosis can be slowed, arrested or even reversed; however, few drugs have been approved specifically for anti-fibrotic treatment. This is in part due to the complex pathways responsible for fibrogenesis and the undesirable side effects of drugs targeting these pathways. Natural products have been utilized for thousands of years as a major component of traditional medicine and currently account for almost one-third of drugs used clinically worldwide. A variety of plant-derived compounds have been demonstrated to have preventative or even reversal effects on fibrosis. This review will discuss the effects and the underlying mechanisms of some of the major plant-derived compounds that have been identified to impact fibrosis.
Collapse
Affiliation(s)
- Patrícia dos Santos Azeredo
- Laboratory of Atherosclerosis, Thrombosis and Cell Therapy, Institute of Biology, State University of Campinas—UNICAMP Campinas, Campinas 13083-970, Brazil;
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| | - E. Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| | - Wayne E. Carver
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| |
Collapse
|
37
|
Hu X, Liu F, Yang H, Qi M, Ren Y, Zhu W, Dai C. Protective Effect and Related Mechanism of Modified Danggui Buxue Decoction on Retinal Oxidative Damage in Mice based on Network Pharmacology. Curr Pharm Des 2024; 30:1912-1926. [PMID: 38835123 DOI: 10.2174/0113816128293824240517113238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 04/01/2024] [Accepted: 04/16/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION Age-related macular degeneration (AMD) is one of the common diseases that cause vision loss in the elderly, and oxidative stress has been considered a major pathogenic factor for AMD. Modified Danggui Buxue Decoction (RRP) has a good therapeutic effect on non-proliferatic diabetic retinopathy and can improve the clinical symptoms of patients. METHODS The key ingredients and core targets of RRP protecting retinal oxidative damage were obtained by Network pharmacology analysis. A mouse retinal oxidative damage model induced by tail vein injection of 1% NaIO3 solution (25 mg/kg) was treated with RRP for 4 weeks and used to verify the pharmacodynamics and related mechanism. AIM This study aimed to predict and verify the protective effect and mechanism of RRP on retinal oxidative damage in mice based on network pharmacology and animal experiments. RESULTS A total of 15 key active components included in RRP interacted with 57 core targets related to retinal oxidative damage (such as AKT1, NFE2L2, HMOX1), mainly involved in the AGE-RAGE signaling pathway in diabetic complications, PI3K-AKT signaling pathway and so on. Further studies in vivo found that RRP improved the retinal oxidative damage, increased the content of SOD and GSH, decreased the content of MDA in mouse serum, promoted the expression of p-PI3K, p-AKT, Nrf2, HO-1 and NQO1 proteins in the mouse retina, and inhibited the expression of Nrf2 in the cytoplasm. CONCLUSION This study revealed that RRP had a protective effect on oxidative damage of the retina in mice, and might exert anti-oxidative effect by activating the PI3K/Akt/Nrf2 signal pathway. This study provided scientific data for the further development of hospital preparations of RRP, and a good theoretical basis for the clinical application of RRP.
Collapse
Affiliation(s)
- Xiangka Hu
- Institute of Materia Medica, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - He Yang
- College of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Mushuang Qi
- College of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ying Ren
- College of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Wanjun Zhu
- College of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chunmei Dai
- Institute of Materia Medica, Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
38
|
Yao Z, Fan Y, Lin L, Kellems RE, Xia Y. Tissue transglutaminase: a multifunctional and multisite regulator in health and disease. Physiol Rev 2024; 104:281-325. [PMID: 37712623 DOI: 10.1152/physrev.00003.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023] Open
Abstract
Tissue transglutaminase (TG2) is a widely distributed multifunctional protein involved in a broad range of cellular and metabolic functions carried out in a variety of cellular compartments. In addition to transamidation, TG2 also functions as a Gα signaling protein, a protein disulfide isomerase (PDI), a protein kinase, and a scaffolding protein. In the nucleus, TG2 modifies histones and transcription factors. The PDI function catalyzes the trimerization and activation of heat shock factor-1 in the nucleus and regulates the oxidation state of several mitochondrial complexes. Cytosolic TG2 modifies proteins by the addition of serotonin or other primary amines and in this way affects cell signaling. Modification of protein-bound glutamines reduces ubiquitin-dependent proteasomal degradation. At the cell membrane, TG2 is associated with G protein-coupled receptors (GPCRs), where it functions in transmembrane signaling. TG2 is also found in the extracellular space, where it functions in protein cross-linking and extracellular matrix stabilization. Of particular importance in transglutaminase research are recent findings concerning the role of TG2 in gene expression, protein homeostasis, cell signaling, autoimmunity, inflammation, and hypoxia. Thus, TG2 performs a multitude of functions in multiple cellular compartments, making it one of the most versatile cellular proteins. Additional evidence links TG2 with multiple human diseases including preeclampsia, hypertension, cardiovascular disease, organ fibrosis, cancer, neurodegenerative diseases, and celiac disease. In conclusion, TG2 provides a multifunctional and multisite response to physiological stress.
Collapse
Affiliation(s)
- Zhouzhou Yao
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yuhua Fan
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Lizhen Lin
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School at Houston, Houston, Texas, United States
| | - Yang Xia
- National Medical Metabolomics International Collaborative Research Center, Central South University, Changsha, Hunan, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
39
|
Xiao Q, Sun CC, Tang CF. Heme oxygenase-1: A potential therapeutic target for improving skeletal muscle atrophy. Exp Gerontol 2023; 184:112335. [PMID: 37984695 DOI: 10.1016/j.exger.2023.112335] [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: 10/07/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Skeletal muscle atrophy is a common muscle disease that is directly caused by an imbalance in protein synthesis and degradation. At the histological level, it is mainly characterized by a reduction in muscle mass and fiber cross-sectional area (CSA). Patients with skeletal muscle atrophy present with reduced motor ability, easy fatigue, and poor life quality. Heme oxygenase-1 (HO-1) is an inducible enzyme that catalyzes the degradation of heme and has attracted much attention for its anti-oxidation effects. In addition, there is growing evidence that HO-1 plays an important role in anti-inflammatory, anti-apoptosis, pro-angiogenesis, and maintaining skeletal muscle homeostasis, making it a potential therapeutic target for improving skeletal muscle atrophy. Here, we review the pathogenesis of skeletal muscle atrophy, the biology of HO-1 and its regulation, and the biological function of HO-1 in skeletal muscle homeostasis, with a specific focus on the role of HO-1 in skeletal muscle atrophy, aiming to observe the therapeutic potential of HO-1 for skeletal muscle atrophy.
Collapse
Affiliation(s)
- Qin Xiao
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan 410012, China; School of Physical Education, Hunan First Normal University, Changsha, Hunan 410205, China
| | - Chen-Chen Sun
- School of Physical Education, Hunan First Normal University, Changsha, Hunan 410205, China.
| | - Chang-Fa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan 410012, China.
| |
Collapse
|
40
|
Xu W, Li F, Zhu L, Cheng M, Cheng Y. Pacenta polypeptide injection alleviates the fibrosis and inflammation in cigarette smoke extracts-induced BEAS-2B cells by modulating MMP-9/TIMP-1 signaling. J Biochem Mol Toxicol 2023; 37:e23453. [PMID: 37437075 DOI: 10.1002/jbt.23453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 05/17/2023] [Accepted: 06/16/2023] [Indexed: 07/14/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) has high morbidity and mortality. Here, we aimed to explore the roles and potential correlation of placenta polypeptide injection (PPI) and MMP-9/TIMP-1 signaling pathway in COPD. BEAS-2B cells were treated with cigarette smoke extract (CSE) to establish a COPD cell model in vitro. The cell survival and cytotoxic effect were measured by CCK-8, LDH release and flow cytometry assays. The inflammatory responses were determined by western blot and ELISA assay. Cell fibrosis was assessed by immunofluorescence and western blot assays. PPI treatment had no cytotoxic effect on BEAS-2B cells until the final concentration reached to 10%. In the range of 0%-8% final concentration, PPI treatment weakened CSE-induced the decrease of cell viability and the increase of LDH level in a concentration-dependent manner. Four percent PPI treatment enhanced cell viability and decreased cell apoptosis of CSE-treated cells in a time-dependent manner. Moreover, 4% PPI treatment significantly decreased inflammatory responses and fibrosis induced by CSE, while AMPA (MMPs agonist) had opposite effects. Notably, AMPA reversed the protective roles of PPI on CSE-induced inflammation and fibrosis. Mechanistically, 4% PPI treatment significantly suppressed MMP-1, MMP-2, MMP-3, MMP-9, MMP-13, and MMP-19 levels, but enhanced TIMP-1, TIMP-2, TIMP-3, and TIMP-4 levels. Among them, MMP-9 and TIMP-1 might be the main target of PPI. PPI effectively attenuated CSE-induced inflammation and fibrosis in vitro by regulating MMP-9/TIMP-1 signaling pathway.
Collapse
Affiliation(s)
- Wei Xu
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, PR China
- Department of Gastroenterology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, PR China
| | - Fuqiang Li
- Department of Gastroenterology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, PR China
| | - Lihong Zhu
- Department of General Practice, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, PR China
| | - Mingliang Cheng
- Department of Infectious Disease, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, PR China
| | - Yiju Cheng
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, PR China
- Suzhou Medical College of Soochow University & The First People's Hospital of Guiyang, Guiyang, Guizhou Province, PR China
| |
Collapse
|
41
|
Li X, Chen D, Ouyang B, Wang S, Li Y, Li L, Zhu S, Zheng G. KLF5/MDM2 Axis Modulates Oxidative Stress and Epithelial-Mesenchymal Transition in Human Lens Epithelial Cells: The Role in Diabetic Cataract. J Transl Med 2023; 103:100226. [PMID: 37532224 DOI: 10.1016/j.labinv.2023.100226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 08/04/2023] Open
Abstract
Diabetic cataract (DC) is a common cause of visual loss in older diabetic subjects. Krüppel-like factor 5 (KLF5) plays an essential role in migration and the epithelial-mesenchymal transition (EMT) in diverse cells and is involved in oxidative stress. However, the effects of KLF5 on DC remain unknown. This study aimed to examine the biological function of KLF5 in DC and its underlying mechanism. The expression patterns of KLF5 were detected in vivo and in vitro. Then, KLF5 was knocked down in human lens epithelial cells (HLECs) to explore its functional roles and underlying mechanisms. Dual-luciferase reporter assay and chromatin immunoprecipitation analysis were used to detect whether KLF5 could bind the promoter of E3 ubiquitin ligase mouse double minute 2 (MDM2), a key regulator of EMT. Lastly, the regulation of KLF5 in the biological behaviors of HLECs via MDM2 was analyzed. We found a significant increase of KLF5 in the DC lens anterior capsular, diabetic rat lens, and high glucose (HG)-stimulated HLECs. Knockdown of KLF5 inhibited oxidative stress, inflammation, migration, and EMT of HG-stimulated HLECs. KLF5 silencing impeded MDM2 expression and restricted the activation of MARK1/FAK and NF-κB signaling pathways in HLECs under HG condition. Additionally, KLF5 was found to bind the MDM2 promoter and enhance the transcriptional activity of MDM2. The protective effects by silencing KLF5 on HG-cultured HLECs could be offset by MDM2 overexpression. We demonstrated that knockdown of KLF5 alleviated oxidative stress, migration, and EMT of HG-cultured HLECs by regulating MDM2, suggesting a potential therapeutic strategy for DC.
Collapse
Affiliation(s)
- Xiao Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Doudou Chen
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Bowen Ouyang
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, Hainan, China
| | - Shengnan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yawei Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Siquan Zhu
- Department of Ophthalmology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Guangying Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
42
|
Jo D, Arjunan A, Choi S, Jung YS, Park J, Jo J, Kim OY, Song J. Oligonol ameliorates liver function and brain function in the 5 × FAD mouse model: transcriptional and cellular analysis. Food Funct 2023; 14:9650-9670. [PMID: 37843873 DOI: 10.1039/d3fo03451h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease worldwide and is accompanied by memory deficits, personality changes, anxiety, depression, and social difficulties. For treatment of AD, many researchers have attempted to find medicinal resources with high effectiveness and without side effects. Oligonol is a low molecular weight polypeptide derived from lychee fruit extract. We investigated the effects of oligonol in 5 × FAD transgenic AD mice, which developed severe amyloid pathology, through behavioral tests (Barnes maze, marble burying, and nestle shredding) and molecular experiments. Oligonol treatment attenuated blood glucose levels and increased the antioxidant response in the livers of 5 × FAD mice. Moreover, the behavioral score data showed improvements in anxiety, depressive behavior, and cognitive impairment following a 2-month course of orally administered oligonol. Oligonol treatment not only altered the circulating levels of cytokines and adipokines in 5 × FAD mice, but also significantly enhanced the mRNA and protein levels of antioxidant enzymes and synaptic plasticity in the brain cortex and hippocampus. Therefore, we highlight the therapeutic potential of oligonol to attenuate neuropsychiatric problems and improve memory deficits in the early stage of AD.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Seoyangro 264, Hwasun 58128, Republic of Korea
| | - Archana Arjunan
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| | - Seoyoon Choi
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Seoyangro 264, Hwasun 58128, Republic of Korea
| | - Yoon Seok Jung
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| | - Jihyun Park
- Department of Food Science and Nutrition, Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea.
- Department of Health Sciences, Graduate School of Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea
| | - Jihoon Jo
- Department of Biomedical Science, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea.
- Department of Health Sciences, Graduate School of Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Seoyangro 264, Hwasun 58128, Republic of Korea
| |
Collapse
|
43
|
Gao Y, Yang Z, He K, Wang Z, Zhang T, Yi J, Zhao L. Voluntary wheel-running improved pulmonary fibrosis by reducing epithelial mesenchymal transformation. Life Sci 2023; 331:122066. [PMID: 37666388 DOI: 10.1016/j.lfs.2023.122066] [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: 06/08/2023] [Revised: 08/15/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
AIMS Pulmonary fibrosis seriously affects the health and life quality of patients. Exercise has been shown to have anti-inflammatory and antioxidant effects, but its effect on pulmonary fibrosis is unclear. In this study, the effect and mechanism of exercise on pulmonary fibrosis induced by paraquat were detected. MAIN METHODS Three data sets were retrieved from GEO data. The biological significance of DEGs generation was determined by GO, KEGG, GSEA, and PPI. Thirty male BALB/C mice were randomly divided into control group, model group and exercise group. H&E staining, Masson staining, Immunohistochemistry and Western blot were used to explore the results. The levels of SOD, CAT, MDA, and GSH in lung tissue were analyzed with detection kits. The levels of inflammatory factors in serum and BALF were measured by ELISA. KEY FINDINGS Compared with the control group, the infiltration of inflammatory cells and fibrotic lesions were increased in the model group. Compared with the model group, voluntary wheel-running reducing the EMT of alveolar epithelial cells, the activation of the Wnt/β-catenin signaling pathway and the level of oxidative distress. Moreover, compared to model group, the serum IL-4, IL-10 and IFN-γ were increased, while the serum CXCL1 were decreased, while the levels of CXCL1, IL-6, IL-10, TNF-α and IFN-γ in the bronchoalveolar lavage fluid were decreased in exercise group. SIGNIFICANCE Voluntary wheel-running reduced inflammatory infiltration and upregulated the expression of antioxidative distress proteins, further to improve the degree of EMT, and ultimately alleviated paraquat induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Yan Gao
- School of Nursing, Jilin University, Changchun 130012, Jilin, China
| | - Zhaoyun Yang
- School of Nursing, Jilin University, Changchun 130012, Jilin, China
| | - Kang He
- School of Nursing, Jilin University, Changchun 130012, Jilin, China
| | - Zeyu Wang
- School of Nursing, Jilin University, Changchun 130012, Jilin, China
| | - Tingyu Zhang
- School of Nursing, Jilin University, Changchun 130012, Jilin, China
| | - Jiang Yi
- Department of Rehabilitation, the Second Hospital of Jilin University, Changchun 130012, Jilin, China.
| | - Lijing Zhao
- School of Nursing, Jilin University, Changchun 130012, Jilin, China.
| |
Collapse
|
44
|
Zou X, Zeng M, Zheng Y, Zheng A, Cui L, Cao W, Wang X, Liu J, Xu J, Feng Z. Comparative Study of Hydroxytyrosol Acetate and Hydroxytyrosol in Activating Phase II Enzymes. Antioxidants (Basel) 2023; 12:1834. [PMID: 37891913 PMCID: PMC10604236 DOI: 10.3390/antiox12101834] [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: 08/02/2023] [Revised: 09/20/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023] Open
Abstract
Nuclear factor E2-related factor 2 (Nrf2) is fundamental to the maintenance of redox homeostasis within cells via the regulation of a series of phase II antioxidant enzymes. The unique olive-derived phenolic compound hydroxytyrosol (HT) is recognized as an Nrf2 activator, but knowledge of the HT derivative hydroxytyrosol acetate (HTac) on Nrf2 activation remains limited. In this study, we observed that an HT pretreatment could protect the cell viability, mitochondrial membrane potential, and redox homeostasis of ARPE-19 cells against a t-butyl hydroperoxide challenge at 50 μM. HTac exhibited similar benefits at 10 μM, indicating a more effective antioxidative capacity compared with HT. HTac consistently and more efficiently activated the expression of Nrf2-regulated phase II enzymes than HT. PI3K/Akt was the key pathway accounting for the beneficial effects of HTac in ARPE-19 cells. A further RNA-Seq analysis revealed that in addition to the consistent upregulation of phase II enzymes, the cells presented distinct expression profiles after HTac and HT treatments. This indicated that HTac could trigger a diverse cellular response despite its similar molecular structure to HT. The evidence in this study suggests that Nrf2 activation is the major cellular activity shared by HTac and HT, and HTac is more efficient at activating the Nrf2 system. This supports its potential future employment in various disease management strategies.
Collapse
Affiliation(s)
- Xuan Zou
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Mengqi Zeng
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Yuan Zheng
- Department of Pediatrics, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Adi Zheng
- School of Medicine, Northwest University, Xi'an 710069, China
| | - Li Cui
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Wenli Cao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xueqiang Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Jiankang Liu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jie Xu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhihui Feng
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| |
Collapse
|
45
|
Hao M, Guan Z, Zhang Z, Ai H, Peng X, Zhou H, Xu J, Gu Q. Atractylodinol prevents pulmonary fibrosis through inhibiting TGF-β receptor 1 recycling by stabilizing vimentin. Mol Ther 2023; 31:3015-3033. [PMID: 37641404 PMCID: PMC10556230 DOI: 10.1016/j.ymthe.2023.08.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 07/11/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023] Open
Abstract
Pirfenidone and nintedanib are only anti-pulmonary fibrosis (PF) drugs approved by the FDA. However, they are not target specific, and unable to modify the disease status. Therefore, it is still desirable to discover more effective agents against PF. Vimentin (VIM) plays key roles in tissue regeneration and wound healing, but its molecular mechanism remains unknown. In this work, we demonstrated that atractylodinol (ATD) significantly inhibits TGF-β1-induced epithelial-mesenchymal transition and fibroblast-to-myofibroblast transition in vitro. ATD also reduces bleomycin-induced lung injury and fibrosis in mice models. Mechanistically, ATD inhibited TGF-β receptor I recycling by binding to VIM (KD = 454 nM) and inducing the formation of filamentous aggregates. In conclusion, we proved that ATD (derived from Atractylodes lancea) modified PF by targeting VIM and inhibiting the TGF-β/Smad signaling pathway. Therefore, VIM is a druggable target and ATD is a proper drug candidate against PF. We prove a novel VIM function that TGF-β receptor I recycling. These findings paved the way to develop new targeted therapeutics against PF.
Collapse
Affiliation(s)
- Mengjiao Hao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Tea Research Institute, Guangdong Academy of Agricultural Sciences, Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China
| | - Zhuoji Guan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhikang Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haopeng Ai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xing Peng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Huihao Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jun Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Qiong Gu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
| |
Collapse
|
46
|
Nie Q, Cao H, Yang J, Liu T, Wang B. Long non-coding RNA NMRAL2P promotes glycolysis and reduces ROS in head and neck tumors by interacting with the ENO1 protein and promoting GPX2 transcription. PeerJ 2023; 11:e16140. [PMID: 37810778 PMCID: PMC10552744 DOI: 10.7717/peerj.16140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Background Metabolic reprogramming is a key marker in the occurrence and development of tumors. This process generates more reactive oxygen species (ROS), promoting the development of oxidative stress. To prevent ROS from harming tumor cells, tumor cells can increase the production of reducing agents to counteract excessive ROS. NMRAL2P has been shown to promote the production of reductive mRNA and plays an important role in the process of oxidative stress. Methods In this study, the clinical data and RNA sequencing of head and neck tumors were obtained from The Cancer Genome Atlas data set. The long non-coding RNA (LncRNA) related to oxidative stress were then identified using differential and correlation analyses. The differential expression and prognosis of the identified lncRNA were then verified using samples from the library of the Second Hospital of Hebei Medical University. Only NMRAL2P was substantially expressed in cancer tissues and predicted a poor prognosis. The tumor-promoting impact of NMRAL2P was then confirmed using in vitro functional assays. The data set was then split into high- and low-expression subgroups based on the median gene expression of NMRAL2P to obtain the mRNA that had a large difference between the two groups, and examine the mechanism of NMRAL2P on GPX2 using quantitative real-time PCR, RNA binding protein immunoprecipitation assay, and chromatin immunoprecipitation. Mass spectrometry was used to identify NMRAL2P-binding proteins and western blotting was used to investigate probable mechanisms. Results The lncRNA NMRAL2P is associated with oxidative stress in head and neck tumors. In vitro functional assays showed that the gene has a cancer-promoting effect, increasing lactic acid and superoxide dismutase production, and reducing the production of ROS and malondialdehyde. NMRAL2P promotes the transcription of GPX2 by binding to transcription factor Nrf2. The gene also inhibits the degradation of ENO1, a crucial enzyme in glycolysis, by binding to protein ENO1. Conclusions This study shows that NMRAL2P can promote glycolysis and reduce the harm to tumor cells caused by ROS. The gene can also be used as a possible target for the treatment of head and neck tumors.
Collapse
Affiliation(s)
- Qian Nie
- Department of Otorhinolaryngology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huan Cao
- Department of Otorhinolaryngology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - JianWang Yang
- Department of Otorhinolaryngology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tao Liu
- Department of Otorhinolaryngology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - BaoShan Wang
- Department of Otorhinolaryngology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
47
|
Tian J, Wang X, Shi H, Wu H, Wang C, Liu N, Guan L, Zhang Z. Sestrin2/Keap1/Nrf2 pathway regulates mucus hypersecretion in pulmonary epithelium induced by traffic-related PM 2.5 and water-soluble extracts. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115455. [PMID: 37708689 DOI: 10.1016/j.ecoenv.2023.115455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 08/18/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
The involvement of fine particulate matter (PM2.5) exposure in the progression of asthma has been extensively discussed in epidemiological and experimental evidence, which aroused widespread attention. Asthma is characterized by mucus hypersecretion. This study investigates the underlying toxic mechanism of traffic-related PM2.5 (TRPM2.5) and water-soluble extracts (WSE) on mucus hypersecretion in the lungs of rats with asthma and 16HBE cells. The ovalbumin-induced rats were administrated by instillation of TRPM2.5 and WSE in the trachea once three days for eight times. The results showed that TRPM2.5 and WSE had an adverse impact on mucus secretion. Specifically, conspicuous mucus stains and increased goblet cells in the bronchial epithelium by PAS staining were found in lung tissues of rats with asthma; MUC5AC gene and protein expression levels in lung tissues of rats with asthma and 16HBE cells were elevated. In addition, TRPM2.5 and WSE triggered oxidative damage via upregulation of malondialdehyde and myeloperoxidase as well as activation of the Sestrin2/Keap1/Nrf2 signaling pathway. Conversely, the knockdown of Sestrin2 effectively inhibited TRPM2.5 and WSE-induced mucus hypersecretion, oxidative stress, and Keap1/Nrf2 signaling pathway and its downstream target gene NQO1. Collectively, it was demonstrated that TRPM2.5 and WSE induced mucus hypersecretion mediated by the Sestrin2/Keap1/Nrf2 pathway.
Collapse
Affiliation(s)
- Jiayu Tian
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Xin Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Yantai Center for Disease Control and Prevention, 264003 Yantai, Shandong, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Hao Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Hongyan Wu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Nannan Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Linlin Guan
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, 56 Xinjian South Road, 030001 Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China.
| |
Collapse
|
48
|
Guo XY, Guo YT, Wang ZR, Jiao YM, Hu Y, Fan LN, Cheng RQ, Qu MM, Zhang C, Song JW, Xu RN, Fan X, Xu W, Zhang JY, Bai BK, Linghu EQ, Chen YK, Ma P, Wang FS. Severe intestinal barrier damage in HIV-infected immunological non-responders. Heliyon 2023; 9:e20790. [PMID: 37876458 PMCID: PMC10590933 DOI: 10.1016/j.heliyon.2023.e20790] [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: 11/04/2022] [Revised: 09/07/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
The intestinal epithelial barrier plays an important role during human immunodeficiency virus (HIV) disease progression. However, the extent to which the intestinal epithelial barrier is damaged in immunological non-responders (INRs) and immunological responders (IRs) is largely unknown. In this study, we investigated and compared the levels of intestinal gland damage and related molecules, including the tight junction protein claudin-1, apoptosis marker caspase-3, HIV DNA, CD4+ T cell count, and inflammation marker tumor necrosis factor-α (TNF-α) among the IRs (n = 10), INRs (n = 8), and healthy controls (HCs, n = 7). Intestinal damage was not completely restored in both INRs and IRs and was more serious in INRs than that in IRs. Moreover, intestinal damage was positively correlated with HIV DNA levels and negatively correlated with CD4+ T cell counts. These results provide insight into understanding the characteristics of intestinal epithelial barrier damage between IRs and INRs.
Collapse
Affiliation(s)
- Xiao-Yan Guo
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yun-Tian Guo
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ze-Rui Wang
- Department of Gastroenterology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yan-Mei Jiao
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yue Hu
- Department of Infectious Diseases, Tianjin Second People's Hospital, Tianjin, China
| | - Li-Na Fan
- Department of Infectious Diseases, Tianjin Second People's Hospital, Tianjin, China
| | | | - Meng-Meng Qu
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chao Zhang
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin-Wen Song
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ruo-Nan Xu
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xing Fan
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Wen Xu
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ji-Yuan Zhang
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Bing-Ke Bai
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - En-Qiang Linghu
- Department of Gastroenterology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yao-Kai Chen
- Department of Infectious Disease, Chongqing Public Health Medical Center, Chongqing, China
| | - Ping Ma
- Department of Infectious Diseases, Tianjin Second People's Hospital, Tianjin, China
| | - Fu-Sheng Wang
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Senior Department of Infectious Diseases, the Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Zhou R, Jin C, Jiao L, Zhang S, Tian M, Liu J, Yang S, Yao W, Zhou F. GGA (geranylgeranylacetone) ameliorates bleomycin-induced lung inflammation and pulmonary fibrosis by inhibiting apoptosis and oxidative stress. Mol Biol Rep 2023; 50:7215-7224. [PMID: 37418084 DOI: 10.1007/s11033-023-08590-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Fibrosis is a response to ongoing cellular injury, disruption, and tissue remodeling, the pathogenesis of which is unknown, and is characterized by extracellular matrix deposition. The antifibrotic effect of Geranylgeranylacetone (GGA), as an inducer of Heat shock protein 70 (HSP70), in liver, kidney and pulmonary fibrosis has been supported by multiple preclinical evidence. However, despite advances in our understanding, the precise roles of HSP70 in fibrosis require further investigation. The purpose of this study was to investigate whether GGA could participate in the progression of pulmonary fibrosis in mice through apoptosis, oxidative stress and inflammation. METHODS AND RESULTS B-cell lymphoma-2(Bcl-2) and Bcl2-Associated X (Bax) are two proteins related to apoptosis. Anti-apoptotic factor Bcl-2 and pro-apoptotic factor Bax are often involved in the apoptotic process in the form of dimer. Immunofluorescence and Western blot results showed that bleomycin (BLM) and transforming growth factor-β (TGF-β) inhibited Bcl-2 expression and promoted Bax expression in vitro and in vivo, respectively. In contrast, GGA treatment reverses this change. Reactive oxygen species (ROS), Malondialdehyde (MDA) and superoxide dismutase (SOD) are markers of oxidative stress, which often reflect oxidative injury of cells. The detection of ROS, MDA and SOD expression showed that TGF-β and BLM treatment could significantly promote oxidative stress, while GGA treatment could alleviate oxidative stress damage. In addition, BLM significantly elevated Tumor necrosis factor-α(TNF-α), Interleukin1β (IL-1β) and Interleukin 6 (IL-6), while scutellarin reversed the above alterations except for that of GGA. RESULTS Taken together, GGA suppressed apoptotic, oxidative stress and inflammation in BLM-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Chaomei Jin
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Linlin Jiao
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Siyu Zhang
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Mei Tian
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Jiamin Liu
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Songtai Yang
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Wu Yao
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| | - Fang Zhou
- Department of Occupational and Environmental Health Science, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| |
Collapse
|
50
|
Yang X, Zhang J, Xiong M, Yang Y, Yang P, Li N, Shi F, Zhu Y, Guo K, Jin Y. NF-κB pathway affects silica nanoparticle-induced fibrosis via inhibited inflammatory response and epithelial-mesenchymal transition in 3D co-culture. Toxicol Lett 2023; 383:141-151. [PMID: 37394155 DOI: 10.1016/j.toxlet.2023.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/06/2023] [Accepted: 06/30/2023] [Indexed: 07/04/2023]
Abstract
Long-term inhalation of silica nanoparticles (SiNPs) can induce pulmonary fibrosis (PF), nevertheless, the potential mechanisms remain elusive. Herein, we constructed a three-dimensional (3D) co-culture model by using Matrigel to investigate the interaction among different cells and potential regulatory mechanisms after SiNPs exposure. Methodologically, we dynamically observed the changes in cell morphology and migration after exposure to SiNPs by co-culturing mouse monocytic macrophages (RAW264.7), human non-small cell lung cancer cells (A549), and medical research council cell strain-5 (MRC-5) in Matrigel for 24 h. Subsequently, we detected the expression of nuclear factor kappa B (NF-κB), inflammatory factor and epithelial-mesenchymal transition (EMT) markers. The results showed that SiNPs produced toxic effects on cells. In the 3D co-culture state, the cell's movement velocity and displacement increased, and the cell migration ability was enhanced. Meanwhile, the expression of inflammatory factor tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) were upregulated, the epithelial marker E-cadherin (E-cad) was downregulated, the mesenchymal marker N-cadherin (N-cad) and myofibroblast marker alpha-smooth muscle actin (α-SMA) expression were upregulated, while NF-κB expression was also upregulated after SiNPs exposure. We further found that cells were more prone to transdifferentiate into myofibroblasts in the 3D co-culture state. Conversely, utilizing the NF-κB-specific inhibitor BAY 11-7082 effectively downregulated the expression of TNF-α, IL-6, interleukin-1β (IL-1β), N-cad, α-SMA, collagen-I (COL I), and fibronectin (FN), the expression of E-cad was upregulated. These findings suggest that NF-κB is involved in regulating SiNPs-induced inflammatory, EMT, and fibrosis in the 3D co-culture state.
Collapse
Affiliation(s)
- Xiaojing Yang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Jing Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Min Xiong
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yushan Yang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Pan Yang
- Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei, China
| | - Ning Li
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Fan Shi
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yaxin Zhu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Keyun Guo
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yulan Jin
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China.
| |
Collapse
|