1
|
Li C, Cao Y, Peng Y, Ma T, Wu F, Hua Y, Wang X, Bai T, Wei Y, Ji P. Changes in PI3K/AKT and NRF2/HO-1 signaling expression and intestinal microbiota in bleomycin-induced pulmonary fibrosis. Food Chem Toxicol 2024; 190:114796. [PMID: 38852756 DOI: 10.1016/j.fct.2024.114796] [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: 01/04/2024] [Revised: 05/19/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Pulmonary fibrosis is the outcome of the prolonged interstitial pneumonia, characterized by excessive accumulation of fibroblasts and collagen deposition, leading to its development. This study aimed to study the changes in PI3K/AKT and NRF2/HO-1 signaling expression and intestinal microbiota in a rat model of a novel bleomycin-induced pulmonary fibrosis. The findings of our study showed the model was successfully established. The results showed that the alveolar septum in the model was significantly widened and infiltrated by severe inflammatory cells. Alveolar atrophy occurred due to the formation of multiple inflammatory foci. During this period, fibrous tissue was distributed in strips and patches, primarily around the pulmonary interstitium and bronchus. Moreover, lung damage and fibrosis progressively worsened over time. The mRNA expression of HO-1 and NRF2 in the model decreased while the mRNA expression of HIF-1α, VEGF, PI3K and AKT increased. Furthermore, it was observed to decrease the protein expression of E-cad, HO-1 and NRF2, and increase the protein expression of α-SMA and p-AKT. Additionally, this model leaded to an imbalance in the intestinal microbiota. This study demonstrate that the novel pulmonary fibrosis model activates the NRF2/HO-1 pathway and the PI3K/AKT pathway in rat lung tissues, and leading to intestinal barrier disorder.
Collapse
Affiliation(s)
- Chenchen Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yuxia Cao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yousheng Peng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Ting Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Fanlin Wu
- Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
| | - Yongli Hua
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiuqin Wang
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry, Yinchuan, 750002, Ningxia, China
| | - Tong Bai
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yanming Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China.
| | - Peng Ji
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China.
| |
Collapse
|
2
|
Tiwari A, Gautam AS, Pandey SK, Singh S, Singh RK. The role of RIPK1 in chronic obstructive pulmonary disease. Drug Discov Today 2024; 29:104020. [PMID: 38740363 DOI: 10.1016/j.drudis.2024.104020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/21/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Necroptosis has emerged as one of the crucial pathological processes involved in the regulation of cell death and inflammation in chronic obstructive pulmonary disease (COPD). Airway epithelial necroptosis is closely linked to COPD pathogenesis. Necroptotic lung cells can release damage-associated molecular patterns (DAMPs) that can initiate a robust inflammatory response. However, the underlying mechanism of necroptosis in COPD is still not clearly understood. Therefore, we aimed to explore the roles and mechanisms of receptor-interacting serine/threonine-protein kinase 1 (RIPK1)-mediated necroptosis in the regulation of inflammatory responses in COPD to provide insights into RIPK1-inhibitor drug discovery efforts and their therapeutic benefits in COPD.
Collapse
Affiliation(s)
- Aman Tiwari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-Raebareli), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India
| | - Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-Raebareli), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India
| | - Shivam Kumar Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-Raebareli), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India
| | - Shreya Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-Raebareli), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-Raebareli), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India.
| |
Collapse
|
3
|
Chen KQ, Wang SZ, Lei HB, Liu X. Necrostatin-1: a promising compound for neurological disorders. Front Cell Neurosci 2024; 18:1408364. [PMID: 38994325 PMCID: PMC11236683 DOI: 10.3389/fncel.2024.1408364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Necrostatin-1, a small molecular alkaloid, was identified as an inhibitor of necroptosis in 2005. Investigating the fundamental mechanism of Necrostatin-1 and its role in various diseases is of great significance for scientific and clinical research. Accumulating evidence suggests that Necrostatin-1 plays a crucial role in numerous neurological disorders. This review aims to provide a comprehensive overview of the potential functions of Necrostatin-1 in various neurological disorders, offering valuable insights for future research.
Collapse
Affiliation(s)
- Ke-Qian Chen
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, University of South China, Hengyang, China
| | - Hai-Bo Lei
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| |
Collapse
|
4
|
He W, Chao J, Gu A, Wang D. Evaluation of 6-PPD quinone toxicity on lung of male BALB/c mice by quantitative proteomics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 922:171220. [PMID: 38412880 DOI: 10.1016/j.scitotenv.2024.171220] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/28/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6-PPDQ), a transformation product of tyre-derived 6-PPD, has been frequently detected in different environments. After 6-PPDQ exposure, we here aimed to examine dynamic lung bioaccumulation, lung injury, and the underlying molecular basis in male BALB/c mice. After single injection at concentration of 4 mg/kg, 6-PPDQ remained in lung up to day 28, and higher level of 6-PPDQ bioaccumulation in lung was observed after repeated injection. Severe inflammation was observed in lung after both single and repeated 6-PPDQ injection as indicated by changes of inflammatory cytokines (TNF-α, IL-6 and IL-10). Sirius red staining and hydroxyproline content analysis indicated that repeated rather than single 6-PPDQ injection induced fibrosis in lung. Repeated 6-PPDQ injection also severely impaired lung function in mice by influencing chord compliance (Cchord) and enhanced pause (Penh). Proteomes analysis was further carried out to identify molecular targets of 6-PPDQ after repeated injection, which was confirmed by transcriptional expression analysis and immunohistochemistry staining. Alterations in Ripk1, Fadd, Il-6st, and Il-16 expressions were identified to be associated with inflammation induction of lung after repeated 6-PPDQ injection. Alteration in Smad2 expression was identified to be associated with fibrosis formation in lung of 6-PPDQ exposed mice. Therefore, long-term and repeated 6-PPDQ exposure potentially resulted in inflammation and fibrosis in lung by affecting certain molecular signals in mammals. Our results suggested several aspects of lung injury caused by 6-PPDQ and provide the underlying molecular basis. These observations implied the possible risks of long-term 6-PPDQ exposure to human health.
Collapse
Affiliation(s)
- Wenmiao He
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China; School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jie Chao
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China
| | - Aihua Gu
- School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Dayong Wang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
5
|
Chen S, Jiang J, Li T, Huang L. PANoptosis: Mechanism and Role in Pulmonary Diseases. Int J Mol Sci 2023; 24:15343. [PMID: 37895022 PMCID: PMC10607352 DOI: 10.3390/ijms242015343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
PANoptosis is a newly defined programmed cell death (PCD) triggered by a series of stimuli, and it engages three well-learned PCD forms (pyroptosis, apoptosis, necroptosis) concomitantly. Normally, cell death is recognized as a strategy to eliminate unnecessary cells, inhibit the proliferation of invaded pathogens and maintain homeostasis; however, vigorous cell death can cause excessive inflammation and tissue damage. Acute lung injury (ALI) and chronic obstructive pulmonary syndrome (COPD) exacerbation is related to several pathogens (e.g., influenza A virus, SARS-CoV-2) known to cause PANoptosis. An understanding of the mechanism and specific regulators may help to address the pathological systems of these diseases. This review presents our understanding of the potential mechanism of PANoptosis and the role of PANoptosis in different pulmonary diseases.
Collapse
Affiliation(s)
| | | | | | - Longshuang Huang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (S.C.); (J.J.); (T.L.)
| |
Collapse
|
6
|
Dorababu A, Maraswami M. Recent Advances (2015-2020) in Drug Discovery for Attenuation of Pulmonary Fibrosis and COPD. Molecules 2023; 28:molecules28093674. [PMID: 37175084 PMCID: PMC10179756 DOI: 10.3390/molecules28093674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/07/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
A condition of scarring of lung tissue due to a wide range of causes (such as environmental pollution, cigarette smoking (CS), lung diseases, some medications, etc.) has been reported as pulmonary fibrosis (PF). This has become a serious problem all over the world due to the lack of efficient drugs for treatment or cure. To date, no drug has been designed that could inhibit fibrosis. However, few medications have been reported to reduce the rate of fibrosis. Meanwhile, ongoing research indicates pulmonary fibrosis can be treated in its initial stages when symptoms are mild. Here, an attempt is made to summarize the recent studies on the effects of various chemical drugs that attenuate PF and increase patients' quality of life. The review is classified based on the nature of the drug molecules, e.g., natural/biomolecule-based, synthetic-molecule-based PF inhibitors, etc. Here, the mechanisms through which the drug molecules attenuate PF are discussed. It is shown that inhibitory molecules can significantly decrease the TGF-β1, profibrotic factors, proteins responsible for inflammation, pro-fibrogenic cytokines, etc., thereby ameliorating the progress of PF. This review may be useful in designing better drugs that could reduce the fibrosis process drastically or even cure the disease to some extent.
Collapse
Affiliation(s)
- Atukuri Dorababu
- Department of Chemistry, SRMPP Government First Grade College, Huvinahadagali 583219, India
| | - Manikantha Maraswami
- Department of Chemistry, Abzena LLC., 360 George Patterson Blvd, Bristol, PA 19007, USA
| |
Collapse
|
7
|
Mitroshina EV, Saviuk M, Vedunova MV. Necroptosis in CNS diseases: Focus on astrocytes. Front Aging Neurosci 2023; 14:1016053. [PMID: 36778591 PMCID: PMC9911465 DOI: 10.3389/fnagi.2022.1016053] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/28/2022] [Indexed: 01/28/2023] Open
Abstract
In the last few years, necroptosis, a recently described type of cell death, has been reported to play an important role in the development of various brain pathologies. Necroptosis is a cell death mechanism that has morphological characteristics similar to necrosis but is mediated by fundamentally different molecular pathways. Necroptosis is initiated by signaling through the interaction of RIP1/RIP3/MLKL proteins (receptor-interacting protein kinase 1/receptor-interacting protein kinase 3/mixed lineage kinase domain-like protein). RIPK1 kinase is usually inactive under physiological conditions. It is activated by stimulation of death receptors (TNFR1, TNFR2, TLR3, and 4, Fas-ligand) by external signals. Phosphorylation of RIPK1 results in the formation of its complex with death receptors. Further, complexes with the second member of the RIP3 and MLKL cascade appear, and the necroptosome is formed. There is enough evidence that necroptosis plays an important role in the pathogenesis of brain ischemia and neurodegenerative diseases. In recent years, a point of view that both neurons and glial cells can play a key role in the development of the central nervous system (CNS) pathologies finds more and more confirmation. Astrocytes play complex roles during neurodegeneration and ischemic brain damage initiating both impair and protective processes. However, the cellular and molecular mechanisms that induce pathogenic activity of astrocytes remain veiled. In this review, we consider these processes in terms of the initiation of necroptosis. On the other hand, it is important to remember that like other types of programmed cell death, necroptosis plays an important role for the organism, as it induces a strong immune response and is involved in the control of cancerogenesis. In this review, we provide an overview of the complex role of necroptosis as an important pathogenetic component of neuronal and astrocyte death in neurodegenerative diseases, epileptogenesis, and ischemic brain damage.
Collapse
|
8
|
Prevention of Bleomycin-Induced Pulmonary Inflammation and Fibrosis in Mice by Bilobalide. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:1973163. [PMID: 36733844 PMCID: PMC9889159 DOI: 10.1155/2023/1973163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/12/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease. Bilobalide (BB) is a sesquiterpene isolated from Ginkgo biloba, and its role in IPF is poorly understood. Mice were intratracheally instilled with 2.5 mg/kg bleomycin (BLM) to induce IPF and then treated with 2.5, 5, and 10 mg/kg BB daily for 21 days. Treatment with BB ameliorated pathological injury and fibrosis of lung tissues in BLM-induced mice. BB suppressed BLM-induced inflammatory response in mice as demonstrated by reduced inflammatory cells counts (leukocytes, neutrophils, macrophages, and lymphocytes) and pro-inflammatory factors (CCL2 and TNF-α), as well as increased CXCL10 levels in BALF. The expression of BLM-induced hydroxyproline, LDH, and pro-fibrotic mediators including fibronectin, collagen I, α-smooth muscle actin (α-SMA), transforming growth factor (TGF)-β1, matrix metalloproteinase (MMP)-2, and MMP-9 in lung tissue was inhibited by BB treatment, and the tissue inhibitor of metalloproteinase-1 (TIMP-1) expression was increased. BB blocked the phosphorylation of JNK and NF-κB, and the nuclear translocation of NF-κB in the lung tissue of mice induced by BLM. Additionally, it abated the activation of NLRP3 inflammasome in lung tissue induced by BLM, which led to the downregulation of IL-18 and IL-1β in BALF. Our present study suggested that BB might ameliorate BLM-induced pulmonary fibrosis by inhibiting the early inflammatory response, which is probably via the inhibition of the JNK/NF-κB/NLRP3 signal pathway. Thus, BB might serve as a therapeutic potential agent for pulmonary inflammation and fibrosis.
Collapse
|
9
|
Elkhoely A, Estfanous RS, Alrobaian M, Borg HM, Kabel AM. Repositioning itraconazole for amelioration of bleomycin-induced pulmonary fibrosis: Targeting HMGB1/TLR4 Axis, NLRP3 inflammasome/NF-κB signaling, and autophagy. Life Sci 2023; 313:121288. [PMID: 36528079 DOI: 10.1016/j.lfs.2022.121288] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/07/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Bleomycin (BLM) is one of the antitumor medications that had proven efficacy in the treatment of a wide range of malignant conditions. Pulmonary fibrosis which is frequently encountered during the course of bleomycin therapy may significantly reduce the potential efficacy of bleomycin in cancer therapy. This study tested the hypothesis that itraconazole may have mitigating effects on BLM-induced pulmonary fibrosis and tried to delineate the potential mechanisms of these effects. MATERIALS AND METHODS In a rat model of pulmonary fibrosis elicited by BLM, the effect of different doses of itraconazole was explored at the biochemical, histopathological, and electron microscopic levels. KEY FINDINGS Itraconazole, in a dose-dependent manner, exhibited significant effects on the pro-oxidant/antioxidant balance, the inflammatory consequences, high-mobility group box 1/toll-like receptor-4 Axis, autophagy and nuclear factor kappa B/Nod-like receptor protein 3 inflammasome signaling and alleviated the histopathological, immunohistochemical, and electron microscopic perturbations induced by BLM in the pulmonary tissues. SIGNIFICANCE In view of the afore-mentioned data, itraconazole may be a promising drug that efficiently mitigates the deleterious effects of BLM on the pulmonary tissues.
Collapse
Affiliation(s)
- Abeer Elkhoely
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Remon S Estfanous
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Majed Alrobaian
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Hany M Borg
- Physiology Department, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh, Egypt
| | - Ahmed M Kabel
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt.
| |
Collapse
|
10
|
Kuai Q, Jian X. Inhibition of miR-23b-3p Ameliorates Scar-Like Phenotypes of Keloid Fibroblasts by Facilitating A20 Expression. Clin Cosmet Investig Dermatol 2022; 15:1549-1559. [PMID: 35967914 PMCID: PMC9365020 DOI: 10.2147/ccid.s367347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/20/2022] [Indexed: 12/16/2022]
Abstract
Purpose Accumulating evidence has reported that microRNAs (miRNAs) play a critical role in the mechanism of keloid formation, and recent research found that miR-23b-3p was upregulated in keloid fibroblasts (KFs). Herein, we explored the potential effect of miR-23b-3p on fibroblasts in keloid. Materials and Methods Clinical tissues, primary KFs and KEL FIB cells were used to detect the expression of miR-23b-3p by performing qRT-PCR. Gene knockdown was carried out to evaluate the molecular and biological changes of primary KFs and KEL FIB cells by conducting CCK-8 assay, flow cytometry and Western blot. The online databases and luciferase reporter assay were utilized to screen and identify the potential target of miR-23b-3p. Results Upregulation of miR-23b-3p was detected in keloid tissues, primary KFs and KF cell line KEL FIB cells, and inhibition of miR-23b-3p promoted apoptosis and suppressed proliferation and the expression of collagen I, collagen III and fibronectin of primary KFs and KEL FIB cells. Further investigation revealed that TNFAIP3, the ubiquitin-editing enzyme A20, was the direct target of miR-23b-3p, and inhibition of miR-23b-3p promoted the expression of A20 in primary KFs and KEL FIB cells. The in vitro assays indicated that A20 suppression inhibited apoptosis and facilitated proliferation and the expression of collagen I, collagen III and fibronectin of miR-23b-3p inhibitor-transfected primary KFs and KEL FIB cells. Finally, we found that miR-23b-3p inhibitor reduced the expression of receptor interacting serine/threonine protein kinase 1 (RIPK1), which was partially reversed by A20 inhibition. Conclusion These findings suggested that inhibition of miR-23b-3p/A20/RIPK1 axis induced apoptosis, limited proliferation and decreased extracellular matrix of KFs, providing a potential therapeutic target for treatment of keloid.
Collapse
Affiliation(s)
- Quan Kuai
- Department of Plastic Surgery, Guangxi Weimei Cosmetology Hospital, Nanning, People’s Republic of China
| | - Xueping Jian
- Department of Oral and Maxillofacial Surgery and Plastic Surgery, the Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People’s Republic of China
- Correspondence: Xueping Jian, Department of Oral and Maxillofacial Surgery and Plastic Surgery, the Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, People’s Republic of China, Tel +86-0791-86362541, Email
| |
Collapse
|
11
|
Liu Y, Chen S, Yu L, Deng Y, Li D, Yu X, Chen D, Lu Y, Liu S, Chen R. Pemafibrate attenuates pulmonary fibrosis by inhibiting myofibroblast differentiation. Int Immunopharmacol 2022; 108:108728. [PMID: 35397395 DOI: 10.1016/j.intimp.2022.108728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/26/2022] [Accepted: 03/18/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Idiopathic pulmonary fibrosis is a chronic progressive disease associated with substantial morbidity and mortality despite advances in medical therapy. Increasing evidence suggests that peroxisome proliferator-activated receptors (PPARs) play important roles in the fibrosis-related diseases and their agonists may become effective therapeutic targets. Pemafibrate is a selective PPARα agonist, but the efficacy against pulmonary fibrosis and mechanisms involved have not been systematically evaluated. Thus, the aims of this study were to explore the role of PPARα in the pulmonary fibrosis and to assess the effect of pemafibrate in vivo and in vitro. METHODS The effects of pemafibrate were evaluated in bleomycin-challenged murine pulmonary fibrosis model and transforming growth factor-beta 1 (TGF-β1) stimulated lung fibroblasts. RESULTS Bleomycin instillation induced body weight loss, declined lung function, pulmonary fibrosis, and extensive collagen deposition in the mice, accompanied with decreased pulmonary expression of PPARα, all of which were partially improved by pemafibrate at a dose of 2 mg/kg. Besides, pemafibrate effectively inhibits TGF-β1-induced myofibroblast differentiation and extracellular matrix (ECM) production in vivo and in vitro. Furthermore, we showed that pemafibrate not only inhibited pulmonary expression of NLRP3 and cleaved caspase-1 in bleomycin-inhaled mice, but also repressed activation of NLRP3/caspase-1 axis in TGF-β1 stimulated lung fibroblasts. CONCLUSION Our data suggest that pemafibrate exerts a marked protection against from the development of pulmonary fibrosis, which could constitute a novel candidate for the treatment for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China; Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shuyu Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China; Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li Yu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Yao Deng
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Difei Li
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Xiu Yu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Dandan Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Ye Lu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China.
| |
Collapse
|
12
|
Utkusavas A, Gurel Gurevin E, Yilmazer N, Uvez A, Oztay F, Bulut H, Ustunova S, Esener OBB, Sonmez K, Erol Kutucu D, Meral I, Dimas K, Armutak EI. Effects of combined administration of doxorubicin and chloroquine on lung pathology in mice with solid Ehrlich ascites carcinoma. Biotech Histochem 2022; 97:555-566. [PMID: 35240890 DOI: 10.1080/10520295.2022.2036369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Combined use of a chemotherapeutic agent and an autophagy inhibitor is a novel cancer treatment strategy. We investigated the effects of chloroquine (CQ) on lung pathology caused by both solid Ehrlich ascites carcinoma (EAC) and doxorubicin (DXR). A control group and eight experimental groups of adult female mice were inoculated subcutaneously with 2.5 × 106 EAC cells. DXR (1.5 mg/kg and 3 mg/kg) and CQ (25 mg/kg and 50 mg/kg) alone or in combination were injected intraperitoneally on days 2, 7 and 12 following inoculation with EAC cells. Lung tissue samples were examined using immunohistochemistry (IHC) for endothelial (eNOS), inducible nitric oxide synthase (iNOS) and neutrophil gelatinase-associated lipocalin (NGAL). Serum catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD) and malondialdehyde (MDA) levels were measured using ELISA. We found decreased levels of iNOS and eNOS in the groups that received 1.5 mg/kg DXR alone and in combination with 25 mg/kg and 50 mg/kg CQ. Combined administration of DXR and CQ partially prevented disruption of alveolar structure. Levels of antioxidant enzymes and MDA were lower in all treated groups; the greatest reduction was observed in mice that received the combination of 25 mg/kg CQ + 1.5 mg/kg DXR. Levels of NGAL were elevated in all treated groups. We found that CQ ameliorated both EAC and DOX induced lung pathology in female mice with solid EAC by reducing oxidative stress.
Collapse
Affiliation(s)
- Ayfer Utkusavas
- Department of Pulmonology, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Ebru Gurel Gurevin
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Nadim Yilmazer
- Department of Biology, Faculty of Arts and Sciences, Tekirdag Namik Kemal University, Tekirdag, Turkey
| | - Ayca Uvez
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Fusun Oztay
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Huri Bulut
- Department of Medical Biochemistry, School of Medicine, Istinye University, Istanbul, Turkey
| | - Savas Ustunova
- Department of Physiology, School of Medicine, Bezmialem Vakıf University, Istanbul, Turkey
| | - Osman B Burak Esener
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Kivilcim Sonmez
- Department of Pathology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Deniz Erol Kutucu
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey.,Department of Biology, Institute of Graduate Studies in Science and Engineering, Istanbul University, Istanbul, Turkey
| | - Ismail Meral
- Department of Physiology, School of Medicine, Bezmialem Vakıf University, Istanbul, Turkey
| | - Konstantinos Dimas
- Department of Pharmacology, School of Medicine, Thessaly University, Larissa, Greece
| | - Elif Ilkay Armutak
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
13
|
Xu Y, Hang WL, Zhou XM, Wu Q. Exploring the Mechanism Whereby Sinensetin Delays the Progression of Pulmonary Fibrosis Based on Network Pharmacology and Pulmonary Fibrosis Models. Front Pharmacol 2021; 12:693061. [PMID: 34220517 PMCID: PMC8249588 DOI: 10.3389/fphar.2021.693061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
The incidence of pulmonary fibrosis (PF), a progressively fatal disease, has increased in recent years. However, there are no effective medicines available. Previous results have shown that sinensetin probably has some curative effects on PF. Therefore, this paper aims to predict the targets of sinensetin using a network pharmacology method and to confirm its effects and functional targets in PF using a mouse PF model. First, network pharmacology analysis showed that sinensetin has 105 functional targets, and 1,698 gene targets closely relate to PF. The intersection of the functional targets and gene targets produced 52 targets for the treatment of PF with sinensetin. The PPIs (protein-protein interactions) led to several potential key target genes, including MAPK1, EGFR, SRC, and PTGS2. The results of GO and KEGG analyses suggested the crucial function of apoptosis in PF and its involvement in the PI3K signaling pathway. Subsequently, we tested the molecular docking of sinensetin with the PI3K protein using the AutoDock4 software. The results showed that sinensetin could fit well into several binding sites of the PI3K protein. Furthermore, we constructed a PF mouse model through one-off intratracheal instillation of bleomycin and then intragastrically administered different concentrations of sinensetin to the model mice. Twenty-eight days later, the mice were sacrificed, and the lung tissues, serum, and bronchoalveolar lavage fluid (BALF) were collected. The in vivo tests showed that the body weight of model mice increased slightly compared with that of PF mice after intragastric sinensetin. HE and Masson staining suggested a certain extent of reduction in the pathology of lung tissues. The expression of collagens I and III, as well as hydroxyproline in the lung tissues, was reduced to a certain extent. IL-6 levels in the serum and BALF decreased markedly. The expression of vimentin and α-SMA in pulmonary tissues decreased. Cell apoptosis, as well as P-PI3K and P-AKT levels, in lung tissues also reduced. In summary, network pharmacology and in vivo test results suggest sinensetin causes an effective delay in the progression of pulmonary fibrosis, and the functional mechanism is likely related to PI3K-AKT signaling.
Collapse
Affiliation(s)
- Yong Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wen-Lu Hang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xian-Mei Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Qi Wu
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
14
|
Zhao X, Zhao B, Zhao Y, Zhang Y, Qian M. Protective effect of anisodamine on bleomycin-induced acute lung injury in immature rats via modulating oxidative stress, inflammation, and cell apoptosis by inhibiting the JAK2/STAT3 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:859. [PMID: 34164493 PMCID: PMC8184412 DOI: 10.21037/atm-21-1750] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Pediatric acute lung injury (ALI) is one of the most common causes of infant mortality. Although lung-protective strategies have developed in recent years, no ALI treatment is currently available. Anisodamine (Ani) is a common drug used to treat gastrointestinal smooth muscle spasm. The protective effects of Ani against acute kidney injury and myocardial injury have been reported. However, the efficacy of Ani on bleomycin (BLM)-induced ALI has not been examined previously. In the present study, we aimed to examine the effects of Ani on bleomycin (BLM)-induced ALI on immature rats. Methods The ALI rat model was established by intratracheally administration of BLM. Ani treatment was performed by an intravenous injection at different concentrations. The lung function of each rat was measured, and then lung tissue structures, apoptosis, and collagen deposition were observed by hematoxylin-eosin staining, terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP nick-end labeling assay, and Masson’s staining, respectively. Enzyme-linked immunosorbent assay was used to detect the levels of inflammatory cytokines. The expression of apoptosis-related proteins and fibrosis-related markers was determined by reverse transcription-polymerase chain reaction and/or Western blot analysis. Finally, the expression levels of Janus tyrosine kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) were determined. Results Our findings indicated that lung function was remarkably decreased in BLM-induced rats, which could be reversed by Ani. Ani treatment increased the levels of antioxidant enzymes, decreased the apoptotic rate and apoptosis-related proteins, and downregulated the expression of fibrosis-related markers. Additionally, Ani treatment also attenuated inflammatory response and suppressed the activation of the JAK2/STAT3 pathway. Conclusions Our results demonstrated that Ani had potent activity against BLM-induced ALI in immature rats through inhibiting the JAK2/STAT3 signaling pathway. Our findings provide supporting evidence to further investigate the therapeutic effect of Ani against ALI in children.
Collapse
Affiliation(s)
- Xiaoqi Zhao
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| | - Bin Zhao
- Department of Neurosurgery, the Second Hospital of Jilin University, Changchun, China
| | - Yinghao Zhao
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, China
| | - Yunfeng Zhang
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| | - Min Qian
- Department of Neonatology, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Jiao B, Tang Y, Liu S, Guo C. Tetrandrine attenuates hyperoxia-induced lung injury in newborn rats via NF-κB p65 and ERK1/2 pathway inhibition. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1018. [PMID: 32953818 PMCID: PMC7475456 DOI: 10.21037/atm-20-5573] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Bronchopulmonary dysplasia (BPD) is an important cause of respiratory illness in preterm newborns that results in significant morbidity and mortality. Hyperoxia is a critical factor in the pathogenesis of BPD, hyperoxia-induced lung injury model has similar pathological manifestations as human BPD. Tetrandrine (Tet) is known to suppress oxidative stress, apoptosis and inflammation. Thus it has been used to prevent organ injuries. However, the protective effect of Tet against hyperoxia-induced lung injury in newborn rats has not been reported. Methods A hyperoxia-induced lung injury model was established using newborn rats exposed to high O2 levels. The models were treated with various concentrations of Tet, and a lung function test was conducted. Then, the lung tissues and blood were collected to detect the effect of Tet on cell apoptosis, inflammatory response, and fibrosis. The effect of Tet on nuclear factor-kappa B (NF-κB) and extracellular signal-regulated kinase1/2 (ERK1/2) pathways was also determined. Results Lung function was decreased in hyperoxia-induced rats, and Tet could reverse this inhibiting effect. For oxidative stress, Tet caused an increase in the levels of antioxidant enzymes. The apoptosis rate and apoptosis-related proteins were decreased in hyperoxia-induced rats after Tet treatment. Additionally, Tet treatment could reduce inflammatory factor levels, while increasing CD4+IFN-γ+ T cell levels and decreasing CD4+IL-4+ T cell levels. Tet treatment was also able to inhibit the expression of fibrosis-related markers and NF-κB and ERK1/2 pathways. Conclusions Tet demonstrated potent activity against hyperoxia-induced lung injury in newborn rats through NF-κB and ERK1/2 pathway inhibition.
Collapse
Affiliation(s)
- Beibei Jiao
- Department of Pediatrics, The first Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Yan Tang
- Department of Pediatrics, The first Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Shan Liu
- Department of Pediatrics, The first Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Chunyan Guo
- Department of Pediatrics, The first Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|