1
|
Huang G, Yang X, Yu Q, Luo Q, Ju C, Zhang B, Chen Y, Liang Z, Xia S, Wang X, Xiang D, Zhong N, Tang XX. Overexpression of STX11 alleviates pulmonary fibrosis by inhibiting fibroblast activation via the PI3K/AKT/mTOR pathway. Signal Transduct Target Ther 2024; 9:306. [PMID: 39523374 PMCID: PMC11551190 DOI: 10.1038/s41392-024-02011-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 09/15/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
Fibroblast activation plays an important role in the occurrence and development of idiopathic pulmonary fibrosis (IPF), which is a progressive, incurable, and fibrotic lung disease. However, the underlying mechanism of fibroblast activation in IPF remains elusive. Here, we showed that the expression levels of STX11 and SNAP25 were downregulated in the lung tissues from patients with IPF and mice with bleomycin (BLM)-induced lung fibrosis as well as in the activated fibroblasts. Upregulation of STX11 or SNAP25 suppressed TGF-β1-induced activation of human lung fibroblasts (HLFs) via promoting autophagy. However, they failed to suppress fibroblast actviation when autophagy was blocked with the use of chloroquine (CQ). In addition, STX11 or SNAP25 could inhibit TGF-β1-induced fibroblast proliferation and migration. In vivo, overexpression of STX11 exerted its protective role in the mice with BLM-induced lung fibrosis. STX11 and SNAP25 mutually promoted expression of each other. Co-IP assay indicated that STX11 has an interaction with SNAP25. Mechanistically, STX11-SNAP25 interaction activated fibroblast autophagy and further inhibited fibroblast activation via blocking the PI3K/AKT/mTOR pathway. Overall, the results suggested that STX11-SNAP25 interaction significantly inhibited lung fibrosis by promoting fibroblast autophagy and suppressing fibroblast activation via blocking the PI3K/ATK/mTOR signaling pathway. Therefore, STX11 serves as a promising therapeutic target in IPF.
Collapse
Affiliation(s)
- Guichuan Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangsheng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingyang Yu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qun Luo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chunrong Ju
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bangyan Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yijing Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zihan Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shu Xia
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaohua Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dong Xiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Laboratory, Bio-island, Guangzhou, China.
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Laboratory, Bio-island, Guangzhou, China.
| |
Collapse
|
2
|
Tang X, Zhu H, Zhou M, Zhang H, Xiao Q, Yuan Q, Sun G, Zhang Z, Chu H. OSGIN1 regulates PM 2.5-induced fibrosis via mediating autophagy in an in vitro model of COPD. Toxicol Lett 2024; 401:35-43. [PMID: 39260748 DOI: 10.1016/j.toxlet.2024.09.003] [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: 09/06/2023] [Revised: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
Fine particulate matter (PM2.5) has been identified as a significant contributing factor to the exacerbation of chronic obstructive pulmonary disease (COPD). It has been observed that PM2.5 may induce lung fibrosis in COPD, although the precise molecular mechanism behind this remains unclear. In a previous study, we demonstrated that PM2.5 upregulates oxidative stress induced growth inhibitor 1 (OSGIN1), which in turn leads to injury in airway epithelial cells, thereby, suggesting a potential link between PM2.5 exposure and COPD. Based on this, we hypothesized that OSGIN1 plays a role in PM2.5-induced fibrosis in COPD. Human bronchial epithelial cells (HBEs) were treated with cigarette smoke extract (CSE) to construct an in vitro model of COPD. Our findings revealed that PM2.5 increased fibrosis indicators and upregulated OSGIN1 in CSE-stimulated HBEs (CSE-HBEs), and knockdown of OSGIN1 reduced the expression of fibrosis indicators. Through the use of microRNA target prediction software and the Gene Expression Omnibus database, we predicted miRNAs that targeted OSGIN1 in COPD. Subsequently, real-time polymerase chain reaction and western blot analysis confirmed that PM2.5 modulated miR-654-5p to regulate OSGIN1 in CSE-HBEs. Western blot demonstrated that OSGIN1 induced autophagy, thereby exacerbating fibrosis in CSE-HBEs. In summary, our results suggest that PM2.5 upregulates OSGIN1 through inhibiting miR-654-5p, leading to increased autophagy and fibrosis in CSE-HBEs.
Collapse
Affiliation(s)
- Xiying Tang
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Huanhuan Zhu
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Meiyu Zhou
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Huilin Zhang
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qi Xiao
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qi Yuan
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Guanting Sun
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhengdong Zhang
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Haiyan Chu
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Yong M, Zhou H, Zeng Y, Yao Y, Zhu H, Hu J. Reduced expression of SMAD7 and consequent reduction of autophagy promotes endometrial stromal-myofibroblast transition and fibrosis. Mol Hum Reprod 2024; 30:gaae036. [PMID: 39412480 DOI: 10.1093/molehr/gaae036] [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: 05/24/2024] [Revised: 09/16/2024] [Indexed: 11/01/2024] Open
Abstract
Abnormal autophagy and the transforming growth factor-β (TGFβ)-SMAD3/7 signaling pathway play an important role in the development of intrauterine adhesions (IUAs); however, the exact underlying mechanisms remain unclear. In this study, we used IUA patient tissue and SMAD7 conditional knockout mice to detect whether SMAD7 effected IUA via regulation of autophagy and the TGFβ-SMAD3 signaling pathway. We applied a combination of techniques for the detection of p-SMAD3, SMAD7, autophagy and fibrosis-related proteins, autophagic flux, and analysis of the SMAD3 binding site. Endometrial tissue of patients with IUA exhibited lower expression levels of SMAD7. In endometrial stromal cells, silencing of SMAD7 inhibited autophagic flux, whereas overexpressed SMAD7 promoted autophagic flux. This SMAD7-mediated autophagic flux regulates the stromal-myofibroblast transition, and these phenotypes were regulated by the TGFβ-SMAD3 signaling pathway. SMAD3 directly binds to the 3'-untranslated region of transcription factor EB (TFEB) and inhibits its transcription. SMAD7 promoted autophagic flux by inhibiting SMAD3, thereby promoting the expression of TFEB. In SMAD7 conditional knockout mice, the endometria showed a fibrotic phenotype. Simultaneously, autophagic flux was inhibited. On administering the autophagy activator rapamycin, this endometrial fibrosis phenotype was partially reversed. The loss of SMAD7 promotes endometrial fibrosis by inhibiting autophagic flux via the TGFβ-SMAD3 pathway. Therefore, this study reveals a potential therapeutic target for IUA.
Collapse
Affiliation(s)
- Min Yong
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Honggui Zhou
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Yuhua Zeng
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Yuqin Yao
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P.R. China
| | - Hongtao Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
4
|
Hu J, Wang N, Jiang Y, Li Y, Qin B, Wang Z, Gao L. BMSCs promote alveolar epithelial cell autophagy to reduce pulmonary fibrosis by inhibiting core fucosylation modifications. Stem Cells 2024; 42:809-820. [PMID: 38982795 DOI: 10.1093/stmcls/sxae044] [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: 01/21/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (PF) is a chronic progressive interstitial lung disease characterized by alveolar epithelial cell (AEC) injury and fibroblast activation. Inadequate autophagy in AECs may result from the activation of several signaling pathways following AEC injury, with glycoproteins serving as key receptor proteins. The core fucosylation (CF) modification in glycoproteins is crucial. Mesenchymal stem cells derived from bone marrow (BMSCs) have the ability to regenerate damaged tissue and treat PF. This study aimed to elucidate the relationship and mechanism of interaction between BMSCs, CF modification, and autophagy in PF. METHODS C57BL/6 male mice, AEC-specific FUT8 conditional knockout (CKO) mice, and MLE12 cells were administered bleomycin (BLM), FUT8 siRNA, and mouse BMSCs, respectively. Experimental techniques including tissue staining, Western blotting, immunofluorescence, autophagic flux detection, and flow cytometry were used in this study. RESULTS First, we found that autophagy was inhibited while FUT8 expression was elevated in PF mice and BLM-induced AEC injury models. Subsequently, CKO mice and MLE12 cells transfected with FUT8 siRNA were used to demonstrate that inhibition of CF modification induces autophagy in AECs and mitigates PF. Finally, mouse BMSCs were used to demonstrate that they alleviate the detrimental autophagy of AECs by inhibiting CF modification and decreasing PF. CONCLUSIONS Suppression of CF modification enhanced the suppression of AEC autophagy and reduced PF in mice. Additionally, through the prevention of CF modification, BMSCs can assist AECs deficient in autophagy and partially alleviate PF.
Collapse
Affiliation(s)
- Jinying Hu
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Nan Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Yu Jiang
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Yina Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Biaojie Qin
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Zhongzhen Wang
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Lili Gao
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| |
Collapse
|
5
|
Lee CE, Kim JY, Yoon JS, Ko J. Role of Inositol-Requiring Enzyme 1 and Autophagy in the Pro-Fibrotic Mechanism Underlying Graves' Orbitopathy. Yonsei Med J 2024; 65:397-405. [PMID: 38910302 PMCID: PMC11199180 DOI: 10.3349/ymj.2023.0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 01/02/2024] [Accepted: 01/25/2024] [Indexed: 06/25/2024] Open
Abstract
PURPOSE Orbital fibroblasts play key roles in the pathogenesis of Graves' orbitopathy (GO), and previous findings have shown that endoplasmic reticulum (ER) stress and autophagy also contribute to GO. In this study, we investigated the presently unclear roles of inositol-requiring enzyme 1 (IRE1) and related autophagy processes in the pro-fibrotic mechanism of GO. MATERIALS AND METHODS Orbital adipose/connective tissues were obtained from eight GO patients and six normal individuals during surgery. GO fibroblasts were transfected with IRE1 small-interfering RNA and treated with bafilomycin A1 (Baf-A1) to evaluate the inhibitory effects of ER stress and autophagy, and protein-expression levels were analyzed through western blotting after stimulation with transforming growth factor (TGF)-β. RESULTS TGF-β stimulation upregulated IRE1 in GO orbital fibroblasts, whereas silencing IRE1 suppressed fibrosis and autophagy responses. Similarly, Baf-A1, an inhibitor of late-phase autophagy, decreased the expression of pro-fibrotic proteins. CONCLUSION IRE1 mediates autophagy and the pro-fibrotic mechanism of GO, which provides a more comprehensive interpretation of GO pathogenesis and suggests potential therapeutic targets.
Collapse
Affiliation(s)
- Chae Eun Lee
- Department of Ophthalmology, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
- Siloam Eye Hospital, Seoul, Korea
| | - Ji-Young Kim
- Department of Ophthalmology, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - JaeSang Ko
- Department of Ophthalmology, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
6
|
Wang Y, He X, Wang H, Hu W, Sun L. Qingfei xieding prescription ameliorates mitochondrial DNA-initiated inflammation in bleomycin-induced pulmonary fibrosis through activating autophagy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117820. [PMID: 38286157 DOI: 10.1016/j.jep.2024.117820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qingfei Xieding prescription was gradually refined and produced by Hangzhou Red Cross Hospital. The raw material includes Ephedra sinica Stapf, Morus alba L., Bombyx Batryticatus, Gypsum Fibrosum, Prunus armeniaca L. var. ansu Maxim., Houttuynia cordata Thunb. , Pueraria edulis Pamp. Paeonia L., Scutellaria baicalensis Georgi and Anemarrhena asphodeloides Bge. It is effective in clinical adjuvant treatment of patients with pulmonary diseases. AIM OF THE STUDY To explore the efficacy and underlying mechanism of Qingfei Xieding (QF) in the treatment of bleomycin-induced mouse model. MATERIALS AND METHODS TGF-β induced fibrotic phenotype in vitro. Bleomycin injection induced lung tissue fibrosis mouse model in vivo. Flow cytometry was used to detect apoptosis, cellular ROS and lipid oxidation. Mitochondria substructure was observed by transmission electron microscopy. Autophagolysosome and nuclear entry of P65 were monitored by immunofluorescence. Quantitative real-time PCR was performed to detect the transcription of genes associated with mtDNA-cGAS-STING pathway and subsequent inflammatory signaling activation. RESULTS TGF-β induced the expression of α-SMA and Collagen I, inhibited cell viability in lung epithelial MLE-12 cells that was reversed by QF-containing serum. TGF-β-mediated downregulation in autophagy, upregulation in lipid oxidation and ROS contents, and mitochondrial damage were rescued by QF-containing serum treatment, but CQ exposure, an autophagy inhibitor, prevented the protective role of QF. In addition to that, the decreased autophagolysosome in TGF-β-exposed MLE-12 cells was reversed by QF and restored to low level in the combination treatment of QF and CQ. Mechanistically, QF-containing serum treatment significantly inhibited mtDNA-cGAS-STING pathway and subsequent inflammatory signaling in TGF-β-challenged cells, which were abolished by CQ-mediated autophagy inhibition. In bleomycin-induced mouse model, QF ameliorated pulmonary fibrosis, reduced mortality, re-activated autophagy in lung tissues and restrained mtDNA-cGAS-STING inflammation pathway. However, the protective effects of QF in bleomycin-induced model mice were also abrogated by CQ. CONCLUSION QF alleviated bleomycin-induced pulmonary fibrosis by activating autophagy, inhibiting mtDNA-cGAS-STING pathway-mediated inflammation. This research recognizes the protection role of QF on bleomycin-induced mouse model, and offers evidence for the potentiality of QF in clinical application for pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Yunguang Wang
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, PR China.
| | - Xinxin He
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, PR China.
| | - Huijie Wang
- Department of Tuberculosis, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, PR China.
| | - Wei Hu
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.
| | - Lifang Sun
- Department of Tuberculosis, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, PR China; Department of Tuberculosis, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
7
|
Huang Z, Cheng S, Jash S, Fierce J, Agudelo A, Higashiyama T, Hanna N, Nakashima A, Saito S, Padbury J, Schuster J, Sharma S. Exploiting sweet relief for preeclampsia by targeting autophagy-lysosomal machinery and proteinopathy. Exp Mol Med 2024; 56:1206-1220. [PMID: 38760513 PMCID: PMC11148015 DOI: 10.1038/s12276-024-01234-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: 08/25/2023] [Revised: 02/02/2024] [Accepted: 02/27/2024] [Indexed: 05/19/2024] Open
Abstract
The etiology of preeclampsia (PE), a severe complication of pregnancy with several clinical manifestations and a high incidence of maternal and fetal morbidity and mortality, remains unclear. This issue is a major hurdle for effective treatment strategies. We recently demonstrated that PE exhibits an Alzheimer-like etiology of impaired autophagy and proteinopathy in the placenta. Targeting of these pathological pathways may be a novel therapeutic strategy for PE. Stimulation of autophagy with the natural disaccharide trehalose and its lacto analog lactotrehalose in hypoxia-exposed primary human trophoblasts restored autophagy, inhibited the accumulation of toxic protein aggregates, and restored the ultrastructural features of autophagosomes and autolysosomes. Importantly, trehalose and lactotrehalose inhibited the onset of PE-like features in a humanized mouse model by normalizing autophagy and inhibiting protein aggregation in the placenta. These disaccharides restored the autophagy-lysosomal biogenesis machinery by increasing nuclear translocation of the master transcriptional regulator TFEB. RNA-seq analysis of the placentas of mice with PE indicated the normalization of the PE-associated transcriptome profile in response to trehalose and lactotrehalose. In summary, our results provide a novel molecular rationale for impaired autophagy and proteinopathy in patients with PE and identify treatment with trehalose and its lacto analog as promising therapeutic options for this severe pregnancy complication.
Collapse
Affiliation(s)
- Zheping Huang
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Shibin Cheng
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Sukanta Jash
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Jamie Fierce
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Anthony Agudelo
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | | | - Nazeeh Hanna
- Division of Neonatology, Department of Pediatrics, New York University Long Island School of Medicine, Mineola, New York, NY, USA
| | - Akitoshi Nakashima
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Shigeru Saito
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - James Padbury
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Jessica Schuster
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA.
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
8
|
Wang S, Yu J, Liu Y, Yu J, Ma Y, Zhou L, Liu X, Liu L, Li W, Niu X. Bletilla striata polysaccharide attenuated the progression of pulmonary fibrosis by inhibiting TGF-β1/Smad signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117680. [PMID: 38171465 DOI: 10.1016/j.jep.2023.117680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/09/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bletilla striata, a traditional medicinal plant, has been utilized as a folk medicine for many years because of its superior biological activity in China. However, Bletilla striata polysaccharide (BSP) has received less attention, and its specific mechanism for ameliorating pulmonary fibrosis is completely unclear. AIMS OF THE STUDY In this study, we aim to assess BSP on the treatment of PF and explore potential mechanisms. MATERIALS AND METHODS BSP was successfully extracted and purified from Bletilla striata. The mechanisms were assessed in bleomycin-induced pulmonary fibrosis model and lung fibroblasts activated by transforming growth factor-β1 (TGF-β1). Histological analysis, immunofluorescence, Western blot and flow cytometry were used to explore the alterations after BSP intervention. RESULTS The results in vivo showed an anti-PF effect of BSP treatment, which reduced pathogenic damages. Furthermore, TGF-β1-induced abnormal migration and upregulated expression of collagen I (COL1A1), vimentin and α-smooth muscle actin (α-SMA) were suppressed by BSP in L929 cells. Moreover, the abnormal proliferation was retarded by inhibiting the cell cycle of G1 to S phase. Immunofluorescence assay showed that BSP activated autophagy and played an antifibrotic role by inhibiting the expression of p62 and phospho-mammalian target of rapamycin (p-mTOR). Last but not least, the suppression of TGF-β1/Smad signaling pathway was critical for BSP to perform therapeutic effects in vitro and in vivo. CONCLUSION The possible mechanisms were involved in improving ECM deposition, regulating cell migration and proliferation, and promoting cellular autophagy. Briefly, all of the above revealed that BSP might be a novel therapy for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yang Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yajing Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lingyi Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
9
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J, Liu O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res 2024; 202:107144. [PMID: 38484858 DOI: 10.1016/j.phrs.2024.107144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Fibrosis is a pathological process that affects multiple organs and is considered one of the major causes of morbidity and mortality in multiple diseases, resulting in an enormous disease burden. Current studies have focused on fibroblasts and myofibroblasts, which directly lead to imbalance in generation and degradation of extracellular matrix (ECM). In recent years, an increasing number of studies have focused on the role of epithelial cells in fibrosis. In some cases, epithelial cells are first exposed to external physicochemical stimuli that may directly drive collagen accumulation in the mesenchyme. In other cases, the source of stimulation is mainly immune cells and some cytokines, and epithelial cells are similarly altered in the process. In this review, we will focus on the multiple dynamic alterations involved in epithelial cells after injury and during fibrogenesis, discuss the association among them, and summarize some therapies targeting changed epithelial cells. Especially, epithelial mesenchymal transition (EMT) is the key central step, which is closely linked to other biological behaviors. Meanwhile, we think studies on disruption of epithelial barrier, epithelial cell death and altered basal stem cell populations and stemness in fibrosis are not appreciated. We believe that therapies targeted epithelial cells can prevent the progress of fibrosis, but not reverse it. The epithelial cell targeting therapies will provide a wonderful preventive and delaying action.
Collapse
Affiliation(s)
- Liuyi Luo
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Oral Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siyao You
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Xinyan Cui
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Qiao Yi
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
10
|
Wu J, Chen Y, Zhang J, Cheng J, Chen Y, Wu T, Zhang M. Inhibition of bleomycin-induced pulmonary fibrosis in SD rats by sea cucumber peptides. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:2876-2887. [PMID: 38018265 DOI: 10.1002/jsfa.13180] [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: 08/14/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is the terminal manifestation of a type of pulmonary disease, which seriously affects the respiratory function of the body, and with no effective cure for treatment. This study evaluated the effect of sea cucumber peptides (SCP) on bleomycin-induced SD rat PF. RESULTS SCP can inhibit the PF induced by bleomycin. PF and SCP did not affect the food intake of rats, but PF reduced the body weight of rats, and SCP could improve the weight loss. SCP reduced lung index in PF rats in a dose-dependent manner. SCP significantly reduced IL-1β, IL-6, TNF-α, α-SMA and VIM expression levels in lung tissue (P < 0.05), significantly decreased TGF-β1 expression level in serum (P < 0.01) and the LSCP group and MSCP group had better inhibitory effects on PF than the HSCP group. Histomorphological results showed that SCP could ameliorate the structural damage of lung tissue, alveolar wall rupture, inflammatory cell infiltration, fibroblast proliferation and deposition of intercellular matrix and collagen fibers caused by PF. The improvement effect of the MSCP group was the most noteworthy in histomorphology. Metabolomics results showed that SCP significantly downregulated catechol, N-acetyl-l-histidine, acetylcarnitine, stearoylcarnitine, d-mannose, l-threonine, l-alanine, glycine, 3-guanidinopropionic acid, prostaglandin D2 and embelic acid d-(-)-β-hydroxybutyric acid expression levels in lung tissue. CONCLUSION SCP ameliorate bleomycin-induced SD rat PF. KEGG pathway analysis proved that SCP intervened in PF mainly via the lysosome pathway, with d-mannose as the key factor. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jianfu Wu
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| | - Yijun Chen
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| | - Jinxuan Zhang
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| | - Jinghuan Cheng
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| | - Yongde Chen
- Research Center for Promoting Optimal Health through Nutritional Intervention, Bestlife Biological Technology Co. Ltd, Tangshan, China
| | - Tao Wu
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| | - Min Zhang
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
- Tianjin Agricultural University, and China-Russia Agricultural Processing Joint Laboratory, Tianjin Agricultural University, Tianjin, China
| |
Collapse
|
11
|
Du J, Yu L, Yang X, Shao F, Xia J, Jin W, Zhang Y, Lei G, Wang Y, Li Y, Zhang J. Regulation of NCOA4-mediated iron recycling ameliorates paraquat-induced lung injury by inhibiting ferroptosis. Cell Commun Signal 2024; 22:146. [PMID: 38388414 PMCID: PMC10885609 DOI: 10.1186/s12964-024-01520-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Paraquat (PQ) is an irreplaceable insecticide in many countries for the advantage of fast-acting and broad-spectrum. However, PQ was classified as the most prevailing poisoning substance for suicide with no specific antidote. Therefore, it is imperative to develop more effective therapeutic agents for the treatment of PQ poisoning. In the present study, both the RNA-Seq and the application of various cell death inhibitors reflected that ferroptosis exerts a crucial regulatory role in PQ poisoning. Moreover, we found PQ strengthens lipid peroxidation as evidenced by different experimental approaches. Of note, pretreatment of iron chelation agent DFO could ameliorate the ferroptotic cell death and alleviate the ferroptosis-related events. Mechanistically, PQ treatment intensively impaired mitochondrial homeostasis, enhanced phosphorylation of AMPK, accelerated the autophagy flux and triggered the activation of Nuclear receptor coactivator 4-ferritin heavy chain (NCOA4-FTH) axis. Importantly, the activation of autophagy was observed prior to the degradation of ferritin, and inhibition of autophagy could inhibit the accumulation of iron caused by the ferritinophagy process. Genetic and pharmacological inhibition of ferritinophagy could alleviate the lethal oxidative events, and rescue the ferroptotic cell death. Excitingly, in the mouse models of PQ poisoning, both the administration of DFO and adeno-associated virus-mediated FTH overexpression significantly reduced PQ-induced ferroptosis and improved the pathological characteristics of pulmonary fibrosis. In summary, the current work provides an in-depth study on the mechanism of PQ intoxication, describes a framework for the further understanding of ferroptosis in PQ-associated biological processes, and demonstrates modulation of iron metabolism may act as a promising therapeutic agent for the management of PQ toxicity.
Collapse
Affiliation(s)
- Jing Du
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xinyi Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fangchun Shao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Weidong Jin
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yinhao Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Guojie Lei
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ying Wang
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
- Department of Clinical Research Center, Luqiao Second People's Hospital, Taizhou, Zhejiang, China.
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
| | - Jun Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
12
|
Jia Q, Ouyang Y, Yang Y, Yao S, Chen X, Hu Z. Osteopontin: A Novel Therapeutic Target for Respiratory Diseases. Lung 2024; 202:25-39. [PMID: 38060060 DOI: 10.1007/s00408-023-00665-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/22/2023] [Indexed: 12/08/2023]
Abstract
Osteopontin (OPN) is a multifunctional phosphorylated protein that is involved in physiological and pathological events. Emerging evidence suggests that OPN also plays a critical role in the pathogenesis of respiratory diseases. OPN can be produced and secreted by various cell types in lungs and overexpression of OPN has been found in acute lung injury/acute respiratory distress syndrome (ALI/ARDS), pulmonary hypertension (PH), pulmonary fibrosis diseases, lung cancer, lung infection, chronic obstructive pulmonary disease (COPD), and asthma. OPN exerts diverse effects on the inflammatory response, immune cell activation, fibrosis and tissue remodeling, and tumorigenesis of these respiratory diseases, and genetic and pharmacological moudulation of OPN exerts therapeutic effects in the treatment of respiratory diseases. In this review, we summarize the recent evidence of multifaceted roles and underlying mechanisms of OPN in these respiratory diseases, and targeting OPN appears to be a potential therapeutic intervention for these diseases.
Collapse
Affiliation(s)
- Qi Jia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| | - Yeling Ouyang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| | - Yiyi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China
| | - Zhiqiang Hu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
13
|
Li WY, Liu JY, Wang ZX, Wang KY, Huang CX, He W, Song JL. Sinapic Acid Attenuates Chronic DSS-Induced Intestinal Fibrosis in C57BL/6J Mice by Modulating NLRP3 Inflammasome Activation and the Autophagy Pathway. ACS OMEGA 2024; 9:1230-1241. [PMID: 38222654 PMCID: PMC10785090 DOI: 10.1021/acsomega.3c07474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/16/2024]
Abstract
Ulcerative colitis (UC) is a chronic gastrointestinal disease that results from repeated inflammation and serious complications. Sinapic acid (SA) is a hydroxycinnamic acid present in a variety of plants that has antioxidant, anti-inflammatory, anticancer, and other protective effects. This study investigated the antifibrotic effect of SA on chronic colitis induced by dextran sulfate sodium salt (DSS) in mice. We observed that SA could significantly reduce clinical symptoms (such as improved body weight loss, increased colon length, and decreased disease activity index score) and pathological changes in mice with chronic colitis. SA supplementation has been demonstrated to repair intestinal mucosal barrier function and maintain epithelial homeostasis by inhibiting activation of the NLRP3 inflammasome and decreasing the expression of IL-6, TNF-α, IL-17A, IL-18, and IL-1β. Furthermore, SA could induce the expression of antioxidant enzymes (Cat, Sod1, Sod2, Mgst1) by activating the Nrf2/keap1 pathway, thus improving antioxidant capacity. Additionally, SA could increase the protein expression of downstream LC3-II/LC3-I and Beclin1 and induce autophagy by regulating the AMPK-Akt/mTOR signaling pathway, thereby reducing the production of intestinal fibrosis-associated proteins Collagen-I and α-SMA. These findings suggest that SA can enhance intestinal antioxidant enzymes, reduce oxidative stress, expedite intestinal epithelial repair, and promote autophagy, thereby ameliorating DSS-induced colitis and intestinal fibrosis.
Collapse
Affiliation(s)
- Wan-Ying Li
- Department
of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin 541100, Guangxi, China
- Department
of Clinical Nutrition, Liuzhou People’s
Hospital, Liuzhou 545006, Guangxi, China
| | - Jun-Yang Liu
- Department
of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin 541100, Guangxi, China
| | - Zi-Xian Wang
- Department
of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin 541100, Guangxi, China
| | - Ke-Ying Wang
- Department
of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin 541100, Guangxi, China
| | - Chun-Xiang Huang
- Department
of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin 541100, Guangxi, China
| | - Wen He
- Guangxi
Key Laboratory of Environmental Exposureomics and Entire Lifecycle
Health, Guilin Medical University, Guilin 541100, Guangxi, China
| | - Jia-Le Song
- Department
of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin 541100, Guangxi, China
- Guangxi
Key Laboratory of Environmental Exposureomics and Entire Lifecycle
Health, Guilin Medical University, Guilin 541100, Guangxi, China
- Department
of Clinical Nutrition and Obstetrics, The
Second Affiliated Hospital of Guilin Medical University, Guilin 541199, Guangxi, China
| |
Collapse
|
14
|
Song C, Xu Z, Liang Q, Mu Y, Liu M, Wu Z, Li X, Li J, Chen H, Wang Y, Gao S, Li A, Yao W, Liu G. OGG1 promoted lung fibrosis by activating fibroblasts via interacting with Snail1. Int Immunopharmacol 2024; 126:111148. [PMID: 37977070 DOI: 10.1016/j.intimp.2023.111148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/01/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
One of abundant DNA lesions induced by reactive oxygen species is 8-oxoguanine (8-oxoG), which compromises genetic instability. 8-oxoG is recognized by the DNA repair protein 8-oxoguanine DNA glycosylase-1 (OGG1) that not only participates in base excision repair but also involves in transcriptional regulation.OGG1 has an important role inIdiopathic Pulmonary Fibrosis (IPF) processing and targeting fibroblasts is a major strategy for the treatment of pulmonary fibrosis, but whether OGG1 activate fibroblast is not clear. In this study, we show that OGG1 expression level is increased at the fibroblast activation stage in mouse lungs induced by bleomycin (BLM) treatment. OGG1 promoted the expression level of fibroblast activation markers (CTGF, fibronectin, and collagen 1) in a pro-fibrotic gene transcriptional regulation pathway via interacting with Snail1, which dependent on 8-oxoG recognition. Global inhibition of OGG1 at the middle stage of lung fibrosis also relieved BLM-induced lung fibrosis in mice. Our results suggest that OGG1 is a target for inhibiting fibroblast activation and a potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Chuge Song
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Respiratory Medicine, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Zhiliang Xu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Breast Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Dongguan Institute of Respiratory Medicine, Guangdong Medical University, Dongguan 523121, China.
| | - Qingyun Liang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Breast Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yifan Mu
- Department of Respiratory Medicine, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Manqi Liu
- Department of Cardiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Zijun Wu
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Xiaomin Li
- Department of Cardiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Jiali Li
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Hongqiao Chen
- Department of Cardiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yahong Wang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Shenglan Gao
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Ao Li
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Weimin Yao
- Department of Respiratory Medicine, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China.
| | - Gang Liu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.
| |
Collapse
|
15
|
Yaribeygi H, Maleki M, Santos RD, Jamialahmadi T, Sahebkar A. Glp-1 Mimetics and Autophagy in Diabetic Milieu: State-of-the-Art. Curr Diabetes Rev 2024; 20:e250124226181. [PMID: 38299271 DOI: 10.2174/0115733998276570231222105959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 02/02/2024]
Abstract
The diabetic milieu is associated with cascades of pathophysiological pathways that culminate in diabetic complications and tissue injuries. Autophagy is an essential process mandatory for cell survival and tissue homeostasis by degrading damaged organelles and removing injured cells. However, it may turn into a pathological process in an aberrant mode in the diabetic and/or malignant milieu. Moreover, autophagy could serve as a promising therapeutic target for many complications related to tissue injury. Glp-1 mimetics are a class of newer antidiabetic agents that reduce blood glucose through several pathways. However, some evidence suggests that they can provide extra glycemic benefits by modulating autophagy, although there is no complete understanding of this mechanism and its underlying molecular pathways. Hence, in the current review, we aimed to provide new insights on the possible impact of Glp-1 mimetics on autophagy and consequent benefits as well as mediating pathways.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Raul D Santos
- Lipid Clinic Heart Institute (Incor), University of São Paulo, Medical School Hospital, São Paulo, Brazil
| | - Tannaz Jamialahmadi
- Medical Toxicolgy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Medical Toxicolgy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Liang C, Bai Y, Miao R, Yang X, Gao L, Liu Y, Zhou J, Guo J, Hu D, Wu J. Celastrol as a candidate drug for silicosis: From bioinformatics and network pharmacology to experimental validation. Int Immunopharmacol 2023; 125:111068. [PMID: 37948856 DOI: 10.1016/j.intimp.2023.111068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
Silicosis, a highly lethal occupational respiratory disease characterized by irreversible pulmonary fibrosis, remains challenging to treat due to its unclear pathogenesis. In this study, bioinformatics, network pharmacology, and experimental validation were combined to explore potential mechanisms and therapeutic drugs for silicosis. First, the differentially expressed genes(DEGs)and pathway enrichment in pulmonary fibrosis were identified by GO and KEGG analysis. Next, the differential genes were submitted to cMap database for drug prediction and celastrol stood out as the most promising candidate drug. Then, network pharmacology analysis identified pharmacological targets of celastrol and demonstrated that celastrol could regulate JAK-STAT, MAPK, and Toll-like receptor signaling pathways. Finally, we verified the therapeutic role and mechanism of celastrol on silicosis. In vivo, celastrol significantly ameliorated CS-induced inflammation and fibrosis in silicosis mice, including inflammatory cell infiltration, collagen fiber and extracellular matrix deposition, fibroblast activation and related factor expression. Moreover, it dramatically improved lung respiratory function of silicosis mice. In vitro, celastrol suppressed CS-induced cytokine expression, apoptosis of macrophages and activation of Stat3 and Erk1/2 signals. Overall, our research identified and verified celastrol as a novel and promising candidate drug for silicosis.
Collapse
Affiliation(s)
- Chao Liang
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China.
| | - Rui Miao
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Xuelian Yang
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Lu Gao
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, Anhui, China; Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, China.
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, Anhui, China; Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, China.
| |
Collapse
|
17
|
Luo W, Gu Y, Fu S, Wang J, Zhang J, Wang Y. Emerging opportunities to treat idiopathic pulmonary fibrosis: Design, discovery, and optimizations of small-molecule drugs targeting fibrogenic pathways. Eur J Med Chem 2023; 260:115762. [PMID: 37683364 DOI: 10.1016/j.ejmech.2023.115762] [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: 06/30/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common fibrotic form of idiopathic diffuse lung disease. Due to limited treatment options, IPF patients suffer from poor survival. About ten years ago, Pirfenidone (Shionogi, 2008; InterMune, 2011) and Nintedanib (Boehringer Ingelheim, 2014) were approved, greatly changing the direction of IPF drug design. However, limited efficacy and side effects indicate that neither can reverse the process of IPF. With insights into the occurrence of IPF, novel targets and agents have been proposed, which have fundamentally changed the treatment of IPF. With the next-generation agents, targeting pro-fibrotic pathways in the epithelial-injury model offers a promising approach. Besides, several next-generation IPF drugs have entered phase II/III clinical trials with encouraging results. Due to the rising IPF treatment requirements, there is an urgent need to completely summarize the mechanisms, targets, problems, and drug design strategies over the past ten years. In this review, we summarize known mechanisms, target types, drug design, and novel technologies of IPF drug discovery, aiming to provide insights into the future development and clinical application of next-generation IPF drugs.
Collapse
Affiliation(s)
- Wenxin Luo
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Siyu Fu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Jifa Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
18
|
Zhao J, Liang Q, Fu C, Cong D, Wang L, Xu X. Autophagy in sepsis-induced acute lung injury: Friend or foe? Cell Signal 2023; 111:110867. [PMID: 37633477 DOI: 10.1016/j.cellsig.2023.110867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening syndrome with high mortality and morbidity, resulting in a heavy burden on family and society. As a key factor that maintains cellular homeostasis, autophagy is regarded as a self-digesting process by which damaged organelles and useless proteins are recycled for cell metabolism, and it thus plays a crucial role during physiological and pathological processes. Recent studies have indicated that autophagy is involved in the pathophysiological process of sepsis-induced ALI, including cell apoptosis, inflammation, and mitochondrial dysfunction, which indicates that regulating autophagy may be beneficial for this disease. However, the role of autophagy in the etiology and treatment of sepsis-induced ALI is not well characterized. This review summarizes the autophagy-related signaling pathways in sepsis-induced ALI, as well as focuses on the dual role of autophagy and its regulation by non-coding RNAs during disease progression, for the development of potential therapeutic strategies in this disease.
Collapse
Affiliation(s)
- Jiayao Zhao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Qun Liang
- Department of Critical Care Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Chenfei Fu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Didi Cong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Long Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiaoxin Xu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
19
|
He J, Wei L, Tan S, Liang B, Liu J, Lu L, Wang T, Wang J, Huang Y, Chen Z, Li H, Zhang L, Zhou Z, Cao Y, Ye X, Yang Z, Xian S, Wang L. Macrophage RAGE deficiency prevents myocardial fibrosis by repressing autophagy-mediated macrophage alternative activation. FASEB J 2023; 37:e23259. [PMID: 37855749 DOI: 10.1096/fj.202300173rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/10/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
Myocardial fibrosis (MF) is the characteristic pathological feature of various cardiovascular diseases that lead to heart failure (HF) or even fatal outcomes. Alternatively, activated macrophages are involved in the development of fibrosis and tissue remodeling. Although the receptor for advanced glycation end products (RAGE) is involved in MF, its potential role in regulating macrophage function in cardiac fibrosis has not been fully investigated. We aimed to determine the role of macrophage RAGE in transverse aortic constriction (TAC)-induced MF. In this study, we found that RAGE expression was markedly increased in the infiltrated alternatively activated macrophages within mice hearts after TAC. RAGE knockout mice showed less infiltration of alternatively activated macrophages and attenuated cardiac hypertrophy and fibrosis compared to the wild-type mice. Our data suggest that mice with macrophage-specific genetic deletion of RAGE were protected from interstitial fibrosis and cardiac dysfunction when subjected to pressure overload, which led to a decreased proportion of alternatively activated macrophages in heart tissues. Our in vitro experiments demonstrated that RAGE deficiency inhibited the differentiation into alternatively activated macrophages by suppressing autophagy activation. In the co-culture system, in vitro polarization of RAW264.7 macrophages toward an alternatively activated phenotype stimulated the expression of α-smooth muscle actin and collagen in cardiac fibroblasts. However, the knockdown of RAGE and inhibition of autophagy in macrophages showed reduced fibroblast-to-myofibroblast transition (FMT). Collectively, our results suggest that RAGE plays an important role in the recruitment and activation of alternatively activated macrophages by regulating autophagy, which contributes to MF. Thus, blockage of RAGE signaling may be an attractive therapeutic target for the treatment of hypertensive heart disease.
Collapse
Affiliation(s)
- Jiaqi He
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Lan Wei
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Shengan Tan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Birong Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Jing Liu
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Lu Lu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, China
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, China
| | - Ting Wang
- Dongguan Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Yusheng Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, China
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, China
| | - Zixin Chen
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, China
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, China
| | - Huan Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, China
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, China
| | - Lu Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Zheng Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhong Cao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Xiaohan Ye
- Dongguan Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqi Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, China
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, China
| | - Shaoxiang Xian
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, China
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, China
| | - Lingjun Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, China
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
20
|
Kaushik S, Bhargava P, Sharma J, Arava S, Nag TC, Arya DS, Bhatia J. Sesamol attenuates bleomycin-induced pulmonary toxicity and fibrosis in experimental animals. J Biochem Mol Toxicol 2023; 37:e23472. [PMID: 37462223 DOI: 10.1002/jbt.23472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 06/29/2023] [Accepted: 07/08/2023] [Indexed: 11/10/2023]
Abstract
Sesamol, a lignan obtained from roasted seeds of Sesamum indicum, has high antioxidant and anti-inflammatory activity. In this study, we have investigated the effect of sesamol on Bleomycin (BLM) induced pulmonary toxicity as well as fibrosis in Wistar rats. Lung toxicity was induced by administration of BLM, 0.015 U/g ip, twice weekly for 28 days whereas lung fibrosis was induced by BLM, 0.015 U/g ip, every 5th day for 49 days. Sesamol administration was started 7 days before first dose of BLM in both the models. It was observed that sesamol 50 mg/kg most effectively attenuated pulmonary toxicity by reducing oxidative stress, inflammation and apoptosis. This dose was further evaluated for its anti-fibrotic effect. It was observed that there was a significant reduction in fibrosis. Lung collagen content was markedly reduced. Furthermore, expression of pro-fibrotic proteins, TGF-β/SMAD and α-SMA, was reduced and that of anti-fibrotic protein, AMPK, was markedly increased. Even though the combination of sesamol with pirfenidone exhibited no additional protection than either drug alone, it is evident from our study that our test drug, sesamol is comparable in efficacy to pirfenidone. Thus, sesamol has promising therapeutic potential in treatment of pulmonary toxicity and fibrosis.
Collapse
Affiliation(s)
- Swati Kaushik
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Poorva Bhargava
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jatin Sharma
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Sudheer Arava
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas C Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir S Arya
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
21
|
Wang Y, Cheng B, Lin YJ, Wang R, Xuan J, Xu HM. Preliminary Study on the Effect and Molecular Mechanism of Tetrandrine in Alleviating Pulmonary Inflammation and Fibrosis Induced by Silicon Dioxide. TOXICS 2023; 11:765. [PMID: 37755775 PMCID: PMC10536946 DOI: 10.3390/toxics11090765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/02/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
This study aims to explore the molecular mechanism of tetrandrine (Tet) in alleviating pulmonary inflammation and fibrosis induced by silica (SiO2) from the perspective of autophagy. C57BL/6J mice were selected as experimental animals, and SiO2 was exposed by intranasal instillation. Tet was intervened by oral gavage. The mice were euthanized on the 7th and 42nd day of SiO2 exposure, and lung tissues were collected for histopathological, molecular biological, immunological, and transmission electron microscopy analysis. The results showed that SiO2 exposure could lead to significant lung inflammation and fibrosis, while Tet could significantly reduce SiO2 exposure-induced lung inflammation and fibrosis. Molecular mechanism research indicated that, compared with SiO2 expose group, Tet intervention could significantly reduce the expression levels of inflammatory cytokines and fibrosis markers (TNF-α, IL-1β, MCP-1, TGF-β1, HYP, Col-I, and Fn), and regulate the expression of key molecules ATG7, microtubule-associated protein 1 light chain 3B (LC3B), and P62 in the autophagy pathway to improve the blocking of autophagic flux, promote the recovery of autophagic lysosomal system function, and inhibit apoptosis. In summary, Tet can alleviate silica-induced lung inflammation and fibrosis, which may be achieved by regulating the expression of key molecules in the autophagy process and associated apoptotic pathway.
Collapse
Affiliation(s)
- Yi Wang
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China
- The Key Laboratory of Environmental Factors and Chronic Disease Control of Ningxia, Yinchuan 750004, China
| | - Bin Cheng
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China
- The Key Laboratory of Environmental Factors and Chronic Disease Control of Ningxia, Yinchuan 750004, China
| | - Yu-Jia Lin
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China
- The Key Laboratory of Environmental Factors and Chronic Disease Control of Ningxia, Yinchuan 750004, China
| | - Rui Wang
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China
- The Key Laboratory of Environmental Factors and Chronic Disease Control of Ningxia, Yinchuan 750004, China
| | - Jie Xuan
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China
- The Fifth People’s Hospital of the Ningxia Hui Autonomous Region, Shizuishan 753000, China
| | - Hai-Ming Xu
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China
- The Key Laboratory of Environmental Factors and Chronic Disease Control of Ningxia, Yinchuan 750004, China
| |
Collapse
|
22
|
Liu Q, Liu X, Wang G, Wu F, Hou Y, Liu H. Genome-wide DNA methylation analysis of Astragalus and Danshen on the intervention of myofibroblast activation in idiopathic pulmonary fibrosis. BMC Pulm Med 2023; 23:325. [PMID: 37667288 PMCID: PMC10478235 DOI: 10.1186/s12890-023-02601-6] [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: 03/30/2023] [Accepted: 08/09/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF), a chronic progressive interstitial lung disease of unknown etiology, is characterized by continuous damage to alveolar epithelial cells, abnormal repair of alveolar tissue, and alveolar wall scar formation. Currently, the recommended treatment for IPF in Western medicine is relatively limited. In contrast, traditional Chinese medicine and compound prescriptions show advantages in the diagnosis and treatment of IPF, which can be attributed to their multi-channel and multi-target characteristics and minimal side-effects. The purpose of this study was to further corroborate the effectiveness and significance of the traditional Chinese medications Astragalus and Danshen in IPF treatment. METHODS We performed whole-genome methylation analysis on nine rat lung tissue samples to determine the epigenetic variation between IPF and non-fibrotic lungs using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses and quantitative reverse transcription polymerase chain reactions. RESULTS We identified differentially methylated regions and 105 associated key functional genes in samples related to IPF and Chinese medicine treatment. Based on the methylation levels and gene expression profiles between the Chinese medicine intervention and pulmonary fibrosis model groups, we speculated that Astragalus and Salvia miltiorrhiza (traditionally known as Danshen) act on the Isl1, forkhead box O3, and Sonic hedgehog genes via regulation at transcriptional and epigenetic levels during IPF. CONCLUSIONS These findings provide novel insights into the epigenetic regulation of IPF, indicate the effectiveness of Astragalus and Danshen in treating IPF, and suggest several promising therapeutic targets for preventing and treating IPF.
Collapse
Affiliation(s)
- Qingyin Liu
- Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, University Science Park, Changqing District, Jinan City, 250355, China
| | - Xue Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369, Jing Shi Road, Jinan City, 250013, China
| | - Guoyu Wang
- Capital Medical University, No. 10, Xizhang Road, Youanmenwai, Fengtai District, Beijing, 100069, China
| | - Fan Wu
- Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, University Science Park, Changqing District, Jinan City, 250355, China
| | - Yuan Hou
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369, Jing Shi Road, Jinan City, 250013, China
| | - Huaman Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369, Jing Shi Road, Jinan City, 250013, China.
| |
Collapse
|
23
|
Lv J, Zhang R, Li D, Liu Y. Resveratrol plays an anti-fibrotic and anti-autophagy role by stimulating miR-192-5p expression in urethral fibrosis. Funct Integr Genomics 2023; 23:241. [PMID: 37450096 DOI: 10.1007/s10142-023-01173-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/25/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Resveratrol (RSV) exerts anti-fibrotic effects on various fibrotic diseases. Whereas the biological role of RSV on urethral fibrosis remains to be elucidated. This study aimed to determine the mechanisms by which RSV affects urethral fibrosis and autophagy. METHODS Sprague‒Dawley rats and primary fibroblasts were treated with transforming growth factor-β1 (TGFβ1) to generate in vivo and in vitro fibrosis models. Then, those were treated with RSV, and autophagy and fibrosis-related indicators were tested. RESULTS Firstly, we found that RSV reversed the upregulation of indicators related to TGFβ1-induced fibrosis (TGFβ1, α-smooth muscle actin, collagen type I, and collagen type III), autophagy (TFEB and LC3), and TGFβR1/Smad4 pathway, as well as the downregulation of p62 and miR-192-5p expression both in vivo and in vitro. Overexpression of miR-192-5p suppressed the upregulation of fibrosis-related markers expression, as well as TFEB and LC3 expression, induced by TGFβ1, while the expression trend of p62 was the opposite. Inhibiting miR-192-5p reversed the effects of RSV on the model group cells. It was also shown that RSV combined with sh-Smad4 inhibited autophagy more effectively than RSV alone. CONCLUSION These results suggest that RSV inhibits urinary fibrosis and autophagy via the miR-192-5p/TGFβR1/Smad4 pathway. RAV may be a potential drug for alleviating urethral fibrosis.
Collapse
Affiliation(s)
- Jin Lv
- Hunan Traditional Chinese Medical College, Zhuzhou, China
| | - Rui Zhang
- Hunan Traditional Chinese Medical College, Zhuzhou, China
| | - DaoYuan Li
- Department of Urology, Hainan Afliated Hospital of Hainan Medical University, Haikou, China
- Department of Urology, Hainan General Hospital, Haikou, China
| | - Yan Liu
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
24
|
Li P, Hao X, Liu J, Zhang Q, Liang Z, Li X, Liu H. miR-29a-3p Regulates Autophagy by Targeting Akt3-Mediated mTOR in SiO 2-Induced Lung Fibrosis. Int J Mol Sci 2023; 24:11440. [PMID: 37511199 PMCID: PMC10380316 DOI: 10.3390/ijms241411440] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/02/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Silicosis is a refractory pneumoconiosis of unknown etiology that is characterized by diffuse lung fibrosis, and microRNA (miRNA) dysregulation is connected to silicosis. Emerging evidence suggests that miRNAs modulate pulmonary fibrosis through autophagy; however, its underlying molecular mechanism remains unclear. In agreement with miRNA microarray analysis, the qRT-PCR results showed that miR-29a-3p was significantly decreased in the pulmonary fibrosis model both in vitro and in vivo. Increased autophagosome was observed via transmission electron microscopy in lung epithelial cell models and lung tissue of silicosis mice. The expression of autophagy-related proteins LC3α/β and Beclin1 were upregulated. The results from using 3-methyladenine, an autophagy inhibitor, or rapamycin, an autophagy inducer, together with TGF-β1, indicated that autophagy attenuates fibrosis by protecting lung epithelial cells. In TGF-β1-treated TC-1 cells, transfection with miR-29a-3p mimics activated protective autophagy and reduced alpha-smooth muscle actin and collagen I expression. miRNA TargetScan predicted, and dual-luciferase reporter experiments identified Akt3 as a direct target of miR-29a-3p. Furthermore, Akt3 expression was significantly elevated in the silicosis mouse model and TGF-β1-treated TC-1 cells. The mammalian target of rapamycin (mTOR) is a central regulator of the autophagy process. Silencing Akt3 inhibited the transduction of the mTOR signaling pathway and activated autophagy in TGF-β1-treated TC-1 cells. These results show that miR-29a-3p overexpression can partially reverse the fibrotic effects by activating autophagy of the pulmonary epithelial cells regulated by the Akt3/mTOR pathway. Therefore, targeting miR-29a-3p may provide a new therapeutic strategy for silica-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Peiyuan Li
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Xiaohui Hao
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan 063210, China
| | - Jiaxin Liu
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Qinxin Zhang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Zixuan Liang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Xinran Li
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan 063210, China
| |
Collapse
|
25
|
Qiu T, Jiang Z, Chen X, Dai Y, Zhao H. Comorbidity of Anxiety and Hypertension: Common Risk Factors and Potential Mechanisms. Int J Hypertens 2023; 2023:9619388. [PMID: 37273529 PMCID: PMC10234733 DOI: 10.1155/2023/9619388] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 06/06/2023] Open
Abstract
Anxiety is more common in patients with hypertension, and these two conditions frequently coexist. Recently, more emphasis has been placed on determining etiology in patients with comorbid hypertension and anxiety. This review focuses on the common risk factors and potential mechanisms of comorbid hypertension and anxiety. Firstly, we analyze the common risk factors of comorbid hypertension and anxiety including age, smoking, alcohol abuse, obesity, lead, and traffic noise. The specific mechanisms underlying hypertension and anxiety were subsequently discussed, including interleukin (IL)-6 (IL-6), IL-17, reactive oxygen species (ROS), and gut dysbiosis. Increased IL-6, IL-17, and ROS accelerate the development of hypertension and anxiety. Gut dysbiosis leads to hypertension and anxiety by reducing short-chain fatty acids, vitamin D, and 5-hydroxytryptamine (5-HT), and increasing trimethylamine N-oxide (TAMO) and MYC. These shared risk factors and potential mechanisms may provide an effective strategy for treating and preventing hypertension and comorbid anxiety.
Collapse
Affiliation(s)
- Tingting Qiu
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The Central Hospital of Changsha City, Hengyang Medical School, University of South China, Changsha, Hunan 410000, China
| | - Zhiming Jiang
- Department of Cardiology, The Fourth Hospital of Changsha, Changsha, Hunan 410006, China
| | - Xuancai Chen
- Urinary Surgery, Affiliated Nanhua Hospital, University of South China, Hengyang 421002, China
| | - Yehua Dai
- Nursing College, University of Xiangnan, Chenzhou, Hunan 423000, China
| | - Hong Zhao
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
26
|
Li R, Kang H, Chen S. From Basic Research to Clinical Practice: Considerations for Treatment Drugs for Silicosis. Int J Mol Sci 2023; 24:ijms24098333. [PMID: 37176040 PMCID: PMC10179659 DOI: 10.3390/ijms24098333] [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: 04/07/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Silicosis, characterized by irreversible pulmonary fibrosis, remains a major global public health problem. Nowadays, cumulative studies are focusing on elucidating the pathogenesis of silicosis in order to identify preventive or therapeutic antifibrotic agents. However, the existing research on the mechanism of silica-dust-induced pulmonary fibrosis is only the tip of the iceberg and lags far behind clinical needs. Idiopathic pulmonary fibrosis (IPF), as a pulmonary fibrosis disease, also has the same problem. In this study, we examined the relationship between silicosis and IPF from the perspective of their pathogenesis and fibrotic characteristics, further discussing current drug research and limitations of clinical application in silicosis. Overall, this review provided novel insights for clinical treatment of silicosis with the hope of bridging the gap between research and practice in silicosis.
Collapse
Affiliation(s)
- Rou Li
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Huimin Kang
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Shi Chen
- Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha 410013, China
| |
Collapse
|
27
|
Han S, Lu Q, Liu X. Advances in cellular senescence in idiopathic pulmonary fibrosis (Review). Exp Ther Med 2023; 25:145. [PMID: 36911379 PMCID: PMC9995810 DOI: 10.3892/etm.2023.11844] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 01/05/2023] [Indexed: 02/17/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible and fatal interstitial lung disease of unknown cause, with a median survival of 2-3 years. Its pathogenesis is unclear and there is currently no effective treatment for IPF. Approximately two-thirds of patients with IPF are >60 years old, with a mean age of 66 years, suggesting a link between aging and IPF. However, the mechanism by which aging promotes development of PF remains unclear. Senescence of alveolar epithelial cells and lung fibroblasts (LFs) and their senescence-associated secretion phenotype (SASP) may be involved in the occurrence and development of IPF. The present review focus on senescence of LFs and epithelial and stem cells, as well as SASP, the activation of profibrotic signaling pathways and potential treatments for pathogenesis of IPF.
Collapse
Affiliation(s)
- Shan Han
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China.,Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Qiangwei Lu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Xiaoqiu Liu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
28
|
Ting L, Feng Y, Zhou Y, Tong Z, Dong Z. IL-27 induces autophagy through regulation of the DNMT1/lncRNA MEG3/ERK/p38 axis to reduce pulmonary fibrosis. Respir Res 2023; 24:67. [PMID: 36869378 PMCID: PMC9985266 DOI: 10.1186/s12931-023-02373-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
PURPOSE Previous studies have shown that interleukin-27 (IL-27) can reduce bleomycin (BLM)-induced pulmonary fibrosis (PF). However, the underlying mechanism by which IL-27 attenuates PF is not fully clear. METHODS In this research, we used BLM to construct a PF mouse model, and MRC-5 cells stimulated by transforming growth factor-β1 (TGF-β1) were used to construct a PF model in vitro. The lung tissue status was observed by Masson and hematoxylin and eosin (HE) staining. To detect gene expression, RT‒qPCR was used. The protein levels were detected by western blotting and immunofluorescence staining. EdU and ELISA were used to detect cell proliferation viability and hydroxyproline (HYP) content, respectively. RESULTS Aberrant IL-27 expression was observed in BLM-induced mouse lung tissues, and the use of IL-27 attenuated mouse lung tissue fibrosis. TGF-β1 induced autophagy inhibition in MRC-5 cells, and IL-27 alleviated MRC-5 cell fibrosis by activating autophagy. The mechanism is inhibition of DNA methyltransferase 1 (DNMT1)-mediated lncRNA MEG3 methylation and ERK/p38 signaling pathway activation. Overexpression of DNMT1, knockdown of lncRNA MEG3, autophagy inhibitor or ERK/p38 signaling pathway inhibitors reversed the positive effect of IL-27 in a lung fibrosis model in vitro. CONCLUSION In conclusion, our study shows that IL-27 upregulates MEG3 expression through inhibition of DNMT1-mediated lncRNA MEG3 promoter methylation, which in turn inhibits ERK/p38 signaling pathway-induced autophagy and attenuates BLM-induced PF, providing a contribution to the elucidation of the potential mechanisms by which IL-27 attenuates PF.
Collapse
Affiliation(s)
- Li Ting
- Department of Respiratory and Critical Care Medicine, Ningbo Huamei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Yingying Feng
- Department of Respiratory and Critical Care Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Ying Zhou
- Department of Respiratory and Critical Care Medicine, Ningbo Huamei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Zhongkai Tong
- Department of Respiratory and Critical Care Medicine, Ningbo Huamei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo Huamei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China.
| |
Collapse
|
29
|
Ahangari F, Price NL, Malik S, Chioccioli M, Bärnthaler T, Adams TS, Kim J, Pradeep SP, Ding S, Cosmos C, Rose KAS, McDonough JE, Aurelien NR, Ibarra G, Omote N, Schupp JC, DeIuliis G, Villalba Nunez JA, Sharma L, Ryu C, Dela Cruz CS, Liu X, Prasse A, Rosas I, Bahal R, Fernández-Hernando C, Kaminski N. microRNA-33 deficiency in macrophages enhances autophagy, improves mitochondrial homeostasis, and protects against lung fibrosis. JCI Insight 2023; 8:e158100. [PMID: 36626225 PMCID: PMC9977502 DOI: 10.1172/jci.insight.158100] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal disease. Recent findings have shown a marked metabolic reprogramming associated with changes in mitochondrial homeostasis and autophagy during pulmonary fibrosis. The microRNA-33 (miR-33) family of microRNAs (miRNAs) encoded within the introns of sterol regulatory element binding protein (SREBP) genes are master regulators of sterol and fatty acid (FA) metabolism. miR-33 controls macrophage immunometabolic response and enhances mitochondrial biogenesis, FA oxidation, and cholesterol efflux. Here, we show that miR-33 levels are increased in bronchoalveolar lavage (BAL) cells isolated from patients with IPF compared with healthy controls. We demonstrate that specific genetic ablation of miR-33 in macrophages protects against bleomycin-induced pulmonary fibrosis. The absence of miR-33 in macrophages improves mitochondrial homeostasis and increases autophagy while decreasing inflammatory response after bleomycin injury. Notably, pharmacological inhibition of miR-33 in macrophages via administration of anti-miR-33 peptide nucleic acids (PNA-33) attenuates fibrosis in different in vivo and ex vivo mice and human models of pulmonary fibrosis. These studies elucidate a major role of miR-33 in macrophages in the regulation of pulmonary fibrosis and uncover a potentially novel therapeutic approach to treat this disease.
Collapse
Affiliation(s)
- Farida Ahangari
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nathan L. Price
- Vascular Biology and Therapeutics Program, Yale Center for Molecular and System Metabolism, Department of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Shipra Malik
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Maurizio Chioccioli
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thomas Bärnthaler
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Taylor S. Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jooyoung Kim
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sai Pallavi Pradeep
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Shuizi Ding
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Cosmos
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kadi-Ann S. Rose
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - John E. McDonough
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nachelle R. Aurelien
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Internal Medicine, Weill Cornell Hospital Medicine, New York, New York, USA
| | - Gabriel Ibarra
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Life Span Medical Group, Department of Internal Medicine, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Norihito Omote
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jonas C. Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Giuseppe DeIuliis
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Julian A. Villalba Nunez
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Charles S. Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xinran Liu
- Center for Cellular and Molecular Imaging (CCMI), Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Antje Prasse
- Department of Pneumology, University of Hannover, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Ivan Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Raman Bahal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale Center for Molecular and System Metabolism, Department of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
30
|
Chen YQ, Gao LD, Liu YL, Shen Y, Diao JL, Yang WH, Wei RL. Autophagy in graves' ophthalmopathy. Front Cell Dev Biol 2023; 11:1158279. [PMID: 37123414 PMCID: PMC10140433 DOI: 10.3389/fcell.2023.1158279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
Graves' ophthalmopathy (GO) is an inflammatory autoimmune disease that affects the eyes. It can significantly alter the quality of life in patients because of its distinctive pathological appearance and the effect on vision. To date, the exact pathological mechanism of GO has not been explicitly discovered. However, several studies have associated autophagy with this disease. Autophagy is a catabolic process that helps maintain homeostasis in all organisms by protecting the cells and tissues from various endogenous and exogenous stress factors. Based on our results, patients affected with GO have comparatively elevated levels of autophagy, which critically affects the pathological mechanism of the GO. In this review, we have summarized the autophagy mechanism in the pathogenesis of GO.
Collapse
Affiliation(s)
- Yu-Qing Chen
- Department of Ophthalmology, Changzheng Hospital of Naval Medicine University, Shanghai, China
| | - Lian-Di Gao
- Department of Nursing, Changzheng Hospital of Naval Medicine University, Shanghai, China
| | - Yi-Lin Liu
- Department of Nursing, Changzheng Hospital of Naval Medicine University, Shanghai, China
| | - Ya Shen
- Department of Ophthalmology, Changzheng Hospital of Naval Medicine University, Shanghai, China
| | - Jia-Le Diao
- Department of Ophthalmology, Changzheng Hospital of Naval Medicine University, Shanghai, China
| | - Wei-Hua Yang
- Shenzhen Eye Hospital, Jinan University, Shenzhen, China
- *Correspondence: Rui-Li Wei, ; Wei-Hua Yang,
| | - Rui-Li Wei
- Department of Ophthalmology, Changzheng Hospital of Naval Medicine University, Shanghai, China
- *Correspondence: Rui-Li Wei, ; Wei-Hua Yang,
| |
Collapse
|
31
|
Liu Y, Li Z, Xiao H, Xie B, He J, Song M, Wang J, Geng J, Dai H, Wang C. USP13 Deficiency Impairs Autophagy and Facilitates Age-related Lung Fibrosis. Am J Respir Cell Mol Biol 2023; 68:49-61. [PMID: 36150040 DOI: 10.1165/rcmb.2022-0002oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an age-related disease. Failure of the proteostasis network with age, including insufficient autophagy, contributes to the pathology of IPF. Mechanisms underlying autophagy disruption in IPF are unclear and may involve regulation of USP (ubiquitin-specific protease) by post-translational modifications. To expand our previous observation of low USP13 expression in IPF, this study evaluated the role of USP13 in age-related lung fibrosis. Here, we demonstrated that Usp13-deficient aged mice exhibited impaired autophagic activity and increased vulnerability to bleomycin-induced fibrosis. Mechanistically, USP13 interacted with and deubiquitinated Beclin 1, and Beclin 1 overexpression abolished the effects of USP13 disruption. In addition, Beclin 1 inhibition resulted in insufficient autophagy and more severe lung fibrosis after bleomycin injury, consistent with the phenotype of aged Usp13-deficient mice. Collectively, we show a protective role of USP13 in age-related pulmonary fibrosis. Aging-mediated USP13 loss impairs autophagic activity and facilitates lung fibrosis through Beclin 1 deubiquitination. Our findings support the notion that age-dependent dysregulation of autophagic regulators enhances vulnerability to lung fibrosis.
Collapse
Affiliation(s)
- Yuan Liu
- Graduate School of Peking Union Medical College and.,National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Zhen Li
- Graduate School of Peking Union Medical College and.,National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Huijuan Xiao
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and.,School of Clinical Medicine, Peking University, Beijing, China
| | - Bingbing Xie
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Jiarui He
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Meiyue Song
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing Geng
- National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Huaping Dai
- Graduate School of Peking Union Medical College and.,National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and
| | - Chen Wang
- Graduate School of Peking Union Medical College and.,State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; and.,School of Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
32
|
Zhang Q, Ban J, Chang S, Qu H, Chen J, Liu F. The aggravate role of exosomal circRNA11:120406118|12040782 on macrophage pyroptosis through miR-30b-5p/NLRP3 axis in silica-induced lung fibrosis. Int Immunopharmacol 2023; 114:109476. [PMID: 36450208 DOI: 10.1016/j.intimp.2022.109476] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/27/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Abstract
Silica dust inhalation could lead to silicosis, and there is no specific biomarker for its early diagnosis and no effective treatment due to the lack of research on its pathogenesis. The homeostasis of macrophages was considered to be crucial during the development of silicosis from persistent chronic inflammation to irreversible fibrosis. However, its regulatory mechanism and the communication between macrophages and others are still not clear. Exosomal circRNAs emerge as favorable candidates for cellular communication. Therefore, our study aimed to illustrate the regulatory mechanism of silicosis from the view of exosomal circRNAs. Our study identified a novel exosomal circRNA, circRNA11:120406118|12040782, in the peripheral serum of silicosis patients. Furthermore, the detailed role of circRNA11:120406118|12040782 was investigated both in silicosis mouse model and in silica-stimulated macrophages and fibroblasts. On the one hand, circRNA11:120406118|12040782 was shown to regulate silica-stimulated macrophage pyroptosis through circRNA11:120406118|12040782/miR-30b-5p/NLRP3 network. And this macrophage-derived cirRNA could promote the activation of fibroblasts. On the other hand, overexpressing miR-30b-5p, the crucial component of circRNA11:120406118|12040782/miR-30b-5p/NLRP3 regulatory network, could inhibit pyroptosis and attenuate silica-induced lung inflammation and fibrosis in mice. Our findings suggested that exosomal circRNA11:120406118|12040782 could aggravate NLRP3-mediated macrophages pyroptosis through sponging miR-30b-5p in silicosis development, which provide an experimental basis and shed light on the early diagnosis and treatment of silicosis.
Collapse
Affiliation(s)
- Qi Zhang
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Jiaqi Ban
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Shuai Chang
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Huiyan Qu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Jie Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China.
| | - Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China.
| |
Collapse
|
33
|
Zhang Y, Zhang J, Fu Z. Role of autophagy in lung diseases and ageing. Eur Respir Rev 2022; 31:31/166/220134. [PMID: 36543345 PMCID: PMC9879344 DOI: 10.1183/16000617.0134-2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2022] Open
Abstract
The lungs face ongoing chemical, mechanical, biological, immunological and xenobiotic stresses over a lifetime. Advancing age progressively impairs lung function. Autophagy is a "housekeeping" survival strategy involved in numerous physiological and pathological processes in all eukaryotic cells. Autophagic activity decreases with age in several species, whereas its basic activity extends throughout the lifespan of most animals. Dysregulation of autophagy has been proven to be closely related to the pathogenesis of several ageing-related pulmonary diseases. This review summarises the role of autophagy in the pathogenesis of pulmonary diseases associated with or occurring in the context of ageing, including acute lung injury, chronic obstructive pulmonary disease, asthma and pulmonary fibrosis, and describes its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China,Corresponding author: Zhiling Fu ()
| |
Collapse
|
34
|
Chen S, Wei Y, Li S, Miao Y, Gu J, Cui Y, Liu Z, Liang J, Wei L, Li X, Zhou H, Yang C. Zanubrutinib attenuates bleomycin-induced pulmonary fibrosis by inhibiting the TGF-β1 signaling pathway. Int Immunopharmacol 2022; 113:109316. [DOI: 10.1016/j.intimp.2022.109316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/11/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
|
35
|
Hu M, Wang Z, Hu X, Wang Y, Wang G, Ding H, Bian M. High-resolution computed tomography diagnosis of pneumoconiosis complicated with pulmonary tuberculosis based on cascading deep supervision U-Net. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 226:107151. [PMID: 36179657 DOI: 10.1016/j.cmpb.2022.107151] [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: 05/30/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE Pulmonary tuberculosis can promote pneumoconiosis deterioration, leading to higher mortality. This study aims to explore the diagnostic value of the cascading deep supervision U-Net (CSNet) model in pneumoconiosis complicated with pulmonary tuberculosis. METHODS A total of 162 patients with pneumoconiosis treated in our hospital were collected as the research objects. Patients were randomly divided into a training set (n = 113) and a test set (n = 49) in proportion (7:3). Based on the high-resolution computed tomography (HRCT), the traditional U-Net, supervision U-Net (SNet), and CSNet prediction models were constructed. Dice similarity coefficients, precision, recall, volumetric overlap error, and relative volume difference were used to evaluate the segmentation model. The area under the receiver operating characteristic curve (AUC) value represents the prediction efficiency of the model. RESULTS There were no statistically significant differences in gender, age, number of positive patients, and dust contact time between patients in the training set and test set (P > 0.05). The segmentation results of CSNet are better than the traditional U-Net model and the SNet model. The AUC value of the CSNet model was 0.947 (95% CI: 0.900∼0.994), which was higher than the traditional U-Net model. CONCLUSION The CSNet based on chest HRCT proposed in this study is superior to the traditional U-Net segmentation method in segmenting pneumoconiosis complicated with pulmonary tuberculosis. It has good prediction efficiency and can provide more clinical diagnostic value.
Collapse
Affiliation(s)
- Maoneng Hu
- Imaging Center, The Third Clinical College of Hefei of Anhui Medical University, The Third People's Hospital of Hefei, Hefei 230022, China.
| | - Zichen Wang
- Imaging Center, The Third Clinical College of Hefei of Anhui Medical University, The Third People's Hospital of Hefei, Hefei 230022, China
| | - Xinxin Hu
- Imaging Center, The Third Clinical College of Hefei of Anhui Medical University, The Third People's Hospital of Hefei, Hefei 230022, China
| | - Yi Wang
- Imaging Center, The Third Clinical College of Hefei of Anhui Medical University, The Third People's Hospital of Hefei, Hefei 230022, China
| | - Guoliang Wang
- Imaging Center, The Third Clinical College of Hefei of Anhui Medical University, The Third People's Hospital of Hefei, Hefei 230022, China
| | - Huanhuan Ding
- Imaging Center, The Third Clinical College of Hefei of Anhui Medical University, The Third People's Hospital of Hefei, Hefei 230022, China
| | - Mingmin Bian
- Imaging Center, The Third Clinical College of Hefei of Anhui Medical University, The Third People's Hospital of Hefei, Hefei 230022, China
| |
Collapse
|
36
|
Liu M, Xue M, Zhang T, Lin R, Guo B, Chen Y, Cheng ZJ, Sun B. Detection of interstitial pneumonia with autoimmune features and idiopathic pulmonary fibrosis are enhanced by involvement of matrix metalloproteinases levels and clinical diagnosis. J Clin Lab Anal 2022; 36:e24734. [PMID: 36250225 DOI: 10.1002/jcla.24734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/06/2022] [Accepted: 10/05/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Higher detection of interstitial pneumonia with autoimmune features (IPAF), and idiopathic pulmonary fibrosis (IPF), has significant diagnostic and therapeutic implications. Some matrix metalloproteinases (MMPs) have become reliable diagnostic biomarkers in IPAF and IPF in previous studies, yet relevant reliability remains to be recognized. MATERIALS AND METHODS In this study, 36 ILDs patients, including 31 IPAF patients (Mean ± SD, 50.20 ± 5.10 years; 16 [51.6%] females) and five IPF patients (Mean ± SD, 61.20 ± 6.73 years; one [20.0%] females) were retrospectively enrolled. Serial serum samples were collected from patients with IPAF and IPF between January 2019 and December 2020. Notably, Serum MMPs levels were measured by U-PLEX Biomarker Group 1(Human) Multiplex Assays (MSD, USA). RESULTS A combination of MMPs and combinatorial biomarkers was strongly associated with clinical subjects in this study (AUC, 0.597 for Stability vs. Improvement and 0.756 for Stability vs. Exacerbation). Importantly, the AUC of MMP-12 reaches 0.730 (p < 0.05, Stability AUC vs. Improvement AUC) while MMP-13 reaches 0.741 (p < 0.05, Stability AUC vs. Exacerbation AUC) showed better performance than other MMPs in two comparisons. CONCLUSIONS Clinical risk factors and MMPs are strongly associated with either stratification of the disease of progression of IPAF or in two IPAF and IPF independent cohorts. To our knowledge, this is the first to illustrate that MMP-12 and MMP-13 may be expected to become typical promising biomarkers in Improvement - IPAF and Exacerbation - IPAF, respectively.
Collapse
Affiliation(s)
- Mingtao Liu
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingshan Xue
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Teng Zhang
- Faculty of Health Sciences, University of Macau, Taipa, China
| | - Runpei Lin
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Baojun Guo
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Zhangkai J Cheng
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Baoqing Sun
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
37
|
Hardouin P, Pible O, Marchandin H, Culotta K, Armengaud J, Chiron R, Grenga L. Quick and wide-range taxonomical repertoire establishment of the cystic fibrosis lung microbiota by tandem mass spectrometry on sputum samples. Front Microbiol 2022; 13:975883. [PMID: 36312921 PMCID: PMC9608366 DOI: 10.3389/fmicb.2022.975883] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/15/2022] [Indexed: 11/19/2022] Open
Abstract
Microorganisms proteotyping by tandem mass spectrometry has been recently shown as a powerful methodology to identify the wide-range taxonomy and biomass of microbiota. Sputum is the recommended specimen for routine microbiological monitoring of Cystic Fibrosis (CF) patients but has been rarely submitted to tandem mass spectrometry-based proteotyping. In this study, we compared the microbial components of spontaneous and induced sputum samples from three cystic fibrosis patients. Although the presence of microbial proteins is much lower than host proteins, we report that the microbiota’s components present in the samples can be identified, as well as host biomarkers and functional insights into the microbiota. No significant difference was found in microorganism abundance between paired spontaneous and induced sputum samples. Microbial proteins linked to resistance, iron uptake, and biofilm-forming ability were observed in sputa independently of the sampling method. This unbiased and enlarged view of the CF microbiome could be highly complementary to culture and relevant for the clinical management of CF patients by improving knowledge about the host-pathogen dynamics and CF pathophysiology.
Collapse
Affiliation(s)
- Pauline Hardouin
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
- Université de Montpellier, Laboratoire Innovations Technologiques pour la Détection et le Diagnostic (Li2D), Bagnols-sur-Cèze, France
| | - Olivier Pible
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
| | - Hélène Marchandin
- HydroSciences Montpellier, CNRS, IRD, Service de Microbiologie et Hygiène Hospitalière, Université de Montpellier, CHU de Nîmes, Nîmes, France
| | - Karen Culotta
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
| | - Jean Armengaud
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
| | - Raphaël Chiron
- HydroSciences Montpellier, CNRS, IRD, Centre de Ressources et de Compétences de la Mucoviscidose, Université de Montpellier, CHU de Montpellier, Montpellier, France
| | - Lucia Grenga
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, Bagnols-sur-Cèze, France
- *Correspondence: Lucia Grenga,
| |
Collapse
|
38
|
Liu H, Pang Q, Cao F, Liu Z, Wei W, Li Z, Long Q, Jiao Y. Number 2 Feibi Recipe Ameliorates Pulmonary Fibrosis by Inducing Autophagy Through the GSK-3β/mTOR Pathway. Front Pharmacol 2022; 13:921209. [PMID: 35903328 PMCID: PMC9315309 DOI: 10.3389/fphar.2022.921209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Number 2 Feibi Recipe (N2FBR) is a traditional Chinese medicine formula for treating idiopathic pulmonary fibrosis. N2FBR inhibits H2O2-mediated oxidative stress damage in alveolar epithelial cells by increasing autophagy, as we previously demonstrated. However, it is unknown if similar mechanisms occur in vivo. We established a pulmonary fibrosis model by instilling bleomycin (BLM) from the airway to examine the effects of N2FBR on pulmonary fibrosis and investigate its probable mechanism in this work. We discovered that N2FBR treatment effectively alleviated interstitial fibrosis as well as collagen deposition, primarily in upregulating SOD, GSH-Px, T-AOC and downregulating MDA content. N2FBR also increased the expression of LC3B, Beclin-1, LAMP1, TFEB and downregulated the expression of p62, legumain. N2FBR treatment boosted the production of autophagosomes, according to the results of the TEM observation. Furthermore, we explored that N2FBR exerted its anti-oxidative stress and pro-autophagy effects via GSK-3β/mTOR signalling pathway. Therefore, these results provide further evidence for the protective effect of N2FBR in pulmonary fibrosis. Our findings could have ramifications for the development of antifibrosis therapies.
Collapse
Affiliation(s)
- Haoge Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qinglu Pang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Fang Cao
- Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhaoheng Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Wan Wei
- Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhipeng Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Long
- Department of Respiratory and Critical Care Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
- *Correspondence: Qi Long, ; Yang Jiao,
| | - Yang Jiao
- Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Qi Long, ; Yang Jiao,
| |
Collapse
|
39
|
Lillehoj EP, Luzina IG, Atamas SP. Mammalian Neuraminidases in Immune-Mediated Diseases: Mucins and Beyond. Front Immunol 2022; 13:883079. [PMID: 35479093 PMCID: PMC9035539 DOI: 10.3389/fimmu.2022.883079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 12/28/2022] Open
Abstract
Mammalian neuraminidases (NEUs), also known as sialidases, are enzymes that cleave off the terminal neuraminic, or sialic, acid resides from the carbohydrate moieties of glycolipids and glycoproteins. A rapidly growing body of literature indicates that in addition to their metabolic functions, NEUs also regulate the activity of their glycoprotein targets. The simple post-translational modification of NEU protein targets-removal of the highly electronegative sialic acid-affects protein folding, alters protein interactions with their ligands, and exposes or covers proteolytic sites. Through such effects, NEUs regulate the downstream processes in which their glycoprotein targets participate. A major target of desialylation by NEUs are mucins (MUCs), and such post-translational modification contributes to regulation of disease processes. In this review, we focus on the regulatory roles of NEU-modified MUCs as coordinators of disease pathogenesis in fibrotic, inflammatory, infectious, and autoimmune diseases. Special attention is placed on the most abundant and best studied NEU1, and its recently discovered important target, mucin-1 (MUC1). The role of the NEU1 - MUC1 axis in disease pathogenesis is discussed, along with regulatory contributions from other MUCs and other pathophysiologically important NEU targets.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Irina G. Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Research Service, Baltimore Veterans Affairs (VA) Medical Center, Baltimore, MD, United States
| | - Sergei P. Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
40
|
Yang G, Tian Y, Li C, Xia J, Qi Y, Yao W, Hao C. LncRNA UCA1 regulates silicosis-related lung epithelial cell-to-mesenchymal transition through competitive adsorption of miR-204-5p. Toxicol Appl Pharmacol 2022; 441:115977. [DOI: 10.1016/j.taap.2022.115977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 11/30/2022]
|
41
|
Yang T, Wang J, Zhao J, Liu Y. Current and prospective applications of exosomal microRNAs in pulmonary fibrosis (Review). Int J Mol Med 2022; 49:37. [PMID: 35088880 PMCID: PMC8815412 DOI: 10.3892/ijmm.2022.5092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022] Open
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive, irreversible and life‑threatening lung disease. However, the pathogenesis and molecular mechanisms of this condition remain unclear. Extracellular vesicles (EVs) are structures derived from the plasma membrane, with a diameter ranging from 30 nm to 5 µm, that play an important role in cell‑to‑cell communications in lung disease, particularly between epithelial cells and the pulmonary microenvironment. In particular, exosomes are a type of EV that can deliver cargo molecules, including endogenous proteins, lipids and nucleic acids, such as microRNAs (miRNAs/miRs). These cargo molecules are encapsulated in lipid bilayers through target cell internalization, receptor‑ligand interactions or lipid membrane fusion. miRNAs are single‑stranded RNA molecules that regulate cell differentiation, proliferation and apoptosis by degrading target mRNAs or inhibiting translation to modulate gene expression. The aim of the present review was to discuss the current knowledge available on exosome biogenesis, composition and isolation methods. The role of miRNAs in the pathogenesis of PF was also reviewed. In addition, emerging diagnostic and therapeutic properties of exosomes and exosomal miRNAs in PF were described, in order to highlight the potential applications of exosomal miRNAs in PF.
Collapse
Affiliation(s)
- Tao Yang
- Department of Respiratory and Critical Care Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
- The First Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Jiaying Zhao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Yang Liu
- Department of Respiratory and Critical Care Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
42
|
Lu Y, Zhong W, Liu Y, Chen W, Zhang J, Zeng Z, Huang H, Qiao Y, Wan X, Meng X, Cai S, Dong H. Anti-PD-L1 antibody alleviates pulmonary fibrosis by inducing autophagy via inhibition of the PI3K/Akt/mTOR pathway. Int Immunopharmacol 2022; 104:108504. [PMID: 35026657 DOI: 10.1016/j.intimp.2021.108504] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/18/2021] [Accepted: 12/26/2021] [Indexed: 11/05/2022]
Abstract
Pulmonary fibrosis is a fatal lung disease for which no effective treatment is available. Previous studies have shown that the expression of programmed cell death-Ligand (PD-L1) is significantly increased in pulmonary fibrosis, and that this is related to the occurrence of this disease. However, the underlying mechanism is not clear. To clarify the efficacy and mechanism of an anti-PD-L1 monoclonal antibody (anti-PD-L1 mAb) as a treatment for pulmonary fibrosis, we conducted histopathological, molecular, and functional analyses in a mouse model of bleomycin-induced pulmonary fibrosis and a cell model of fibrosis induced by transforming growth factor-beta 1 (TGF-β1). Our results indicate that PD-L1 is highly expressed in the lung fibrosis model. The anti-PD-L1 mAb significantly alleviated bleomycin-induced lung structural disorders and collagen deposition in mice and inhibited the proliferation, migration, activation and extracellular matrix deposition of TGF-β1-induced lung fibroblasts. Interestingly, the anti-PD-L1 mAb could also alleviate the autophagy impairment observed in pulmonary fibrosis. The potential mechanism is through the downregulation of the PI3K/Akt/mTOR signaling pathway. Our study provides evidence of the crucial ability of anti-PD-L1 mAbs to activate autophagy in the context of pulmonary fibrosis, providing a new strategy for the treatment of this disease.
Collapse
Affiliation(s)
- Ye Lu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Wenshan Zhong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yuanyuan Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Weimou Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jinming Zhang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Zhaojin Zeng
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Haohua Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yujie Qiao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xuan Wan
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xiaojing Meng
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
43
|
Wu ZX, Huang X, Cai MJ, Huang PD, Guan Z. Development and Validation of a Prognostic Index Based on Genes Participating in Autophagy in Patients With Lung Adenocarcinoma. Front Oncol 2022; 11:799759. [PMID: 35145906 PMCID: PMC8821527 DOI: 10.3389/fonc.2021.799759] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
BackgroundLung adenocarcinoma (LUAD) is a deadly respiratory system malignancy with poor prognosis. Autophagy is essential for the beginning, development, and therapy resistance of cancer. However, the expression of genes participating in autophagy in LUAD and their associations with prognosis remain unclear.MethodsPredictive genes participating in autophagy in LUAD samples from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets were investigated. TCGA and GEO cohorts were divided into two risk groups, while the low-risk group having a longer overall survival (OS) time. This article aims to point out the interaction between genes participating in autophagy and immune function, immune checkpoints, and m6a in LUAD. The prediction model was designed for exploring least absolute shrinkage and selection operator (LASSO) regression. It has been revealed that gene expression and autophagy are inextricably connected.ResultsGenes participating in autophagy were shown to be somewhat overexpressed in the high-risk group even though no different clinical symptoms were present, indicating that they might be used in a model to predict LUAD prognosis. The majority of genes participating in autophagy prognostic signatures controlled immunological and tumor-related pathways, according to gene set enrichment analysis (GSEA). KRT6A, KYNU, IGFBP1, DKK1, PKP2, PLEK2, GAPDH, FLNC, and NTSR1 might be related to the oncology process for LUAD patients. CERS4, CMAHP, and PLEKHB1 have been identified as being associated with low risk in patients with LUAD. Furthermore, the immune function and m6a gene expression differed significantly between the two groups.ConclusionsGenes participating in autophagy are connected to the development and progression of LUAD. LUAD patients’ prognoses are often foreseen utilizing matched prognostic models. Genes participating in autophagy in LUAD may be therapeutic targets that ought to be investigated more.
Collapse
Affiliation(s)
- Zi-Xuan Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuyan Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Pei-Dong Huang
- Yunnan University of Chinese Medicine, Kunming, China
- *Correspondence: Pei-Dong Huang,
| | - Zunhui Guan
- Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
44
|
Gao Q, Chang X, Yang M, Zheng J, Gong X, Liu H, Li K, Wang X, Zhan H, Li S, Feng S, Sun X, Sun Y. LncRNA MEG3 restrained pulmonary fibrosis induced by NiO NPs via regulating hedgehog signaling pathway-mediated autophagy. ENVIRONMENTAL TOXICOLOGY 2022; 37:79-91. [PMID: 34608745 DOI: 10.1002/tox.23379] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
Long noncoding RNA maternally expressed gene 3 (lncRNA MEG3) was down-regulated in pulmonary fibrosis of rats induced by Nickel oxide nanoparticles (NiO NPs), while the downstream regulatory mechanisms of MEG3 remain unclear. This study aimed to investigate the relationship among MEG3, Hedgehog (Hh) signaling pathway and autophagy in pulmonary fibrosis caused by NiO NPs. The pulmonary fibrosis model in rats was constructed by intratracheal instillation of 0.015, 0.06, and 0.24 mg/kg NiO NPs twice a week for 9 weeks. Collagen deposition model was established by treating A549 cells with 25, 50, and 100 μg/mL NiO NPs for 24 h. Our results indicated that NiO NPs activated Hh pathway, down-regulated the expression of MEG3, and reduced autophagy activity in vivo and in vitro. Meanwhile, the autophagy process was promoted by Hh pathway inhibitor (CDG-0449), while the collagen formation in A549 cells was reduced by autophagy activator (Rapamycin). Furthermore, the overexpressed MEG3 inhibited the activation of Hh pathway, resulting in autophagy activity enhancement along with collagen formation reduction. In summary, lncRNA MEG3 can restrain pulmonary fibrosis induced by NiO NPs via regulating hedgehog signaling pathway-mediated autophagy, which may serve as a potential therapeutic strategy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Qing Gao
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Mengmeng Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Jinfa Zheng
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xuefeng Gong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Han Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Kun Li
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiaoxia Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Haibing Zhan
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Sheng Li
- Department of Public Health, The First People's Hospital of Lanzhou city, Lanzhou, China
| | - Sanwei Feng
- Institute of Occupational Diseases, Gansu Baoshihua Hospital, Lanzhou, China
| | - Xingchang Sun
- Institute of Occupational Diseases, Gansu Baoshihua Hospital, Lanzhou, China
| | - Yingbiao Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
45
|
Fagone E, Fruciano M, Gili E, Sambataro G, Vancheri C. Developing PI3K Inhibitors for Respiratory Diseases. Curr Top Microbiol Immunol 2022; 436:437-466. [DOI: 10.1007/978-3-031-06566-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
46
|
Abstract
Herbal compounds including those already well-established in traditional Chinese medicine have been increasingly tested in the treatment of various diseases. Recent studies have shown that herbal compounds can be of benefit also for pulmonary silicosis as they can diminish changes associated with silica-induced inflammation, fibrosis, and oxidative stress. Due to a lack of effective therapeutic strategies, development of novel approaches which may be introduced particularly in the early stage of the disease, is urgently needed. This review summarizes positive effects of several alternative plant-based drugs in the models of experimental silicosis with a potential for subsequent clinical investigation and use in future.
Collapse
Affiliation(s)
- J Adamcakova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | |
Collapse
|
47
|
Abstract
Herbal compounds including those already well-established in traditional Chinese medicine have been increasingly tested in the treatment of various diseases. Recent studies have shown that herbal compounds can be of benefit also for pulmonary silicosis as they can diminish changes associated with silica-induced inflammation, fibrosis, and oxidative stress. Due to a lack of effective therapeutic strategies, development of novel approaches which may be introduced particularly in the early stage of the disease, is urgently needed. This review summarizes positive effects of several alternative plant-based drugs in the models of experimental silicosis with a potential for subsequent clinical investigation and use in future.
Collapse
Affiliation(s)
- J ADAMCAKOVA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - D MOKRA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| |
Collapse
|
48
|
What is a Therapeutic Potential of N-Acetylcysteine in Lung Silicosis? ACTA MEDICA MARTINIANA 2021. [DOI: 10.2478/acm-2021-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Lung silicosis is a serious pulmonary disease caused by an exposure of lung to inhaled silicon dioxide (SiO2) or silica. Although pathomechanisms of the disease have not been fully elucidated, oxidative stress has been recognized as a fundamental factor triggering a fibrotizing inflammation leading to irreversible changes in lung tissue. Based on this knowledge, therapeutic potential of various antioxidants has been intensively discussed. Among them, N-acetylcysteine with its multiple anti-inflammatory and antioxidant actions and a long-term experience with its clinical use in various diseases appears as a very promising choice. The purpose of this article is to review the therapeutic effects of N-acetylcysteine particularly in relation to a lung injury and to point out a potential of N-acetylcysteine in the treatment of lung silicosis.
Collapse
|
49
|
Abstract
Pulmonary fibrosis is the end stage of a broad range of heterogeneous interstitial lung diseases and more than 200 factors contribute to it. In recent years, the relationship between virus infection and pulmonary fibrosis is getting more and more attention, especially after the outbreak of SARS-CoV-2 in 2019, however, the mechanisms underlying the virus-induced pulmonary fibrosis are not fully understood. Here, we review the relationship between pulmonary fibrosis and several viruses such as Human T-cell leukemia virus (HTLV), Human immunodeficiency virus (HIV), Cytomegalovirus (CMV), Epstein–Barr virus (EBV), Murine γ-herpesvirus 68 (MHV-68), Influenza virus, Avian influenza virus, Middle East Respiratory Syndrome (MERS)-CoV, Severe acute respiratory syndrome (SARS)-CoV and SARS-CoV-2 as well as the mechanisms underlying the virus infection induced pulmonary fibrosis. This may shed new light on the potential targets for anti-fibrotic therapy to treat pulmonary fibrosis induced by viruses including SARS-CoV-2.
Collapse
Affiliation(s)
- Wei Jie Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China. .,Guangzhou Laboratory, Bio-island, Guangzhou, China.
| |
Collapse
|
50
|
Wang X, Liu T, Huang Y, Dai Y, Lin H. Regulation of transforming growth factor-β signalling by SUMOylation and its role in fibrosis. Open Biol 2021; 11:210043. [PMID: 34753319 PMCID: PMC8580444 DOI: 10.1098/rsob.210043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is an abnormal healing process that only repairs the structure of an organ after injury and does not address damaged functions. The pathogenesis of fibrosis is multifactorial and highly complex; numerous signalling pathways are involved in this process, with the transforming growth factor-β (TGF-β) signalling pathway playing a central role. TGF-β regulates the generation of myofibroblasts and the epithelial-mesenchymal transition by regulating transcription and translation of downstream genes and precisely regulating fibrogenesis. The TGF-β signalling pathway can be modulated by various post-translational modifications, of which SUMOylation has been shown to play a key role. In this review, we focus on the function of SUMOylation in canonical and non-canonical TGF-β signalling and its role in fibrosis, providing promising therapeutic strategies for fibrosis.
Collapse
Affiliation(s)
- Xinyi Wang
- First Clinical Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Ting Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Yifei Huang
- First Clinical Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Yifeng Dai
- Second Clinical Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| |
Collapse
|