1
|
Gao L, Zeng X, Huang Y, Huang L. Baicalin inhibits LPS-induced apoptosis and inflammation in WI- 38 cells by promoting FOXA2/TRIM27 Interaction: Implications for pediatric pneumonia mechanisms. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04162-3. [PMID: 40266299 DOI: 10.1007/s00210-025-04162-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Pediatric pneumonia is lung inflammation in newborns caused by many factors, which can impair the respiratory, circulatory and nervous systems and affect their growth and development. Baicalin, a flavonoid separated from Scutellaria baicalensis Georgi, possesses anti-inflammatory effects in lung diseases. The aim of this study was to explore the molecular mechanism of baicalin in exerting a protective effect in neonatal pneumonia. METHODS Effect of baicalin on viability of human fibroblast cells (WI-38 cell) was detected by CCK-8 assay. Then, the WI-38 cells were treated with lipopolysaccharide (LPS). Cell apoptosis and inflammatory cytokines were assessed by flow cytometry and ELISA. Additionally, the oxidative stress and endoplasmic reticulum stress (ERS) were evaluated using specific assays. The mRNA and protein levels were assessed by qRT-PCR and western blot. Finally, the binding between FOXA2 and TRIM27 was predicted and verified by employing the Jaspar database, ChIP and dual luciferase reporter assays. RESULTS 1-40 µM baicalin had no impact on the viability in WI-38 cells, and 40 µM baicalin increased the viability of LPS-inhibited cells. Besides, baicalin mitigated the effects of LPS on apoptosis, inflammation, oxidative stress and ERS in WI-38 cells. Moreover, TRIM27 exhibited low expression levels in pediatric pneumonia and LPS-induced cells. Furthermore, baicalin promoted TRIM27 expression and inhibited the effects of LPS induction on cell production. Mechanically, FOXA2 was positively correlated with TRIM27 expression and baicalin inhibited the adverse effects of LPS induction on WI-38 cells via FOXA2/TRIM27. CONCLUSION These findings suggested that baicalin miaght exert protective effects against pediatric pneumonia by modulating FOXA2/TRIM27-dependent pathways.
Collapse
Affiliation(s)
- Lihua Gao
- Department of Paediatrics, South Hospital of Ganzhou People's Hospital, Pediatric Doctor's Office, 4th Floor, Pediatric Building, No. 16 Meiguan Avenue, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| | - Xiaojin Zeng
- Department of Paediatrics, Ruijin Maternal and Child Health Hospital, Ruijin City, 341000, Jiangxi Province, China
| | - Yubo Huang
- Department of Paediatrics, South Hospital of Ganzhou People's Hospital, Pediatric Doctor's Office, 4th Floor, Pediatric Building, No. 16 Meiguan Avenue, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| | - Leming Huang
- Department of Paediatrics, South Hospital of Ganzhou People's Hospital, Pediatric Doctor's Office, 4th Floor, Pediatric Building, No. 16 Meiguan Avenue, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China.
| |
Collapse
|
2
|
Cheng Y, Jiang M, Qin X, Mao J, Liu Y, Zhu G. Baicalin attenuates LPS-induced periodontal inflammation response by inhibiting autophagy. BMC Oral Health 2025; 25:513. [PMID: 40211339 PMCID: PMC11987269 DOI: 10.1186/s12903-025-05913-7] [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: 01/18/2025] [Accepted: 03/31/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Periodontal disease causes gradual damage to the periodontal ligament and alveolar bone, ultimately resulting in tooth loss. This condition This condition results from the intricate interaction between bacterial infections and the host's inflammatory responses, driving disease progression. Autophagy, an essential process for cellular balance under stress, plays a vital role in the response to periodontal pathogens. Baicalin (BA), a flavonoid extracted from Scutellaria baicalensis, is recognized for its potent anti-inflammatory effects. However, its influence on autophagy in periodontal health is not fully characterized, representing a vital gap in therapeutic understanding. PURPOSE This study investigates the therapeutic potential of BA in periodontal disease by examining its regulatory effects on autophagy and inflammation in PDLCs. METHODS Periodontal ligament cells (PDLCs) were exposed to various concentrations of BA, and cell proliferation was measured using the CCK-8 assay. Anti-inflammatory responses were analyzed by quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA). Autophagy levels were quantified using immunofluorescence, transmission electron microscopy (TEM), and Western blotting. To identify potential targets of BA, an integrated approach combining network pharmacology and RNA sequencing (RNA-Seq) was employed. These analyses were subsequently validated using qRT-PCR, molecular docking and dynamics simulations. RESULTS BA significantly reduced lipopolysaccharide (LPS)-induced inflammatory responses in PDLCs, as evidenced by a decrease in the levels of interleukin (IL)-1β and IL-6. RNA-Seq analysis indicated that these effects were associated with autophagy-related processes. Notably, BA decreased Beclin-1 levels, reduced the LC3BII/I ratio, diminished LC3B protein staining, and decreased the number of autophagosomes. Furthermore, BA triggered the activation of the PI3K/AKT/mTOR pathway, demonstrated by the increased phosphorylation of these proteins. CONCLUSION BA acts as a protective agent against LPS-induced periodontal inflammation by modulating autophagy, positioning it as a promising candidate for future periodontal therapies.
Collapse
Affiliation(s)
- Yifan Cheng
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan, 430030, Hubei, China
| | - Ming Jiang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan, 430030, Hubei, China
| | - Xu Qin
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan, 430030, Hubei, China
| | - Jing Mao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan, 430030, Hubei, China.
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital for Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, China.
| | - Guangxun Zhu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan, 430030, Hubei, China.
| |
Collapse
|
3
|
Zhang Y, Wang L, Zeng J, Shen W. Research advances in polyphenols from Chinese herbal medicine for the prevention and treatment of chronic obstructive pulmonary disease: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03945-y. [PMID: 40035820 DOI: 10.1007/s00210-025-03945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is a global health problem due to its high death and morbidity worldwide, which is characterized by an incompletely reversible limitation in airflow that is not fully reversible. Unfortunately, Western medical treatments are unable to reverse the progressive decline in lung function. Importantly, polyphenolic compounds isolated from Chinese herbal medicine exhibited therapeutic/interventional effects on COPD in preclinical studies. This review systematically analyzed the pathogenesis of COPD, such as inflammation, oxidative stress, protease/antiprotease imbalance, aging, cell death, and dysbiosis of gut microbiota. Moreover, this review summarized the regulatory mechanisms of natural polyphenolic compounds for the treatment of COPD. Several studies have demonstrated that natural polyphenolic compounds have therapeutic effects on COPD by regulating various biological processes, such as anti-inflammatory, reduction of oxidative damage, anti-cell death, and inhibition of airway hyperglycemia. Mechanistically, this review found that the promising effects of natural polyphenolic compounds on COPD were mainly achieved through modulating the NF-κB and MAPK inflammatory pathways, the Nrf2 oxidative stress pathway, and the SIRT1/PGC-1α lung injury pathway. Furthermore, this review analyzed the efficacy and safety of natural polyphenolic compounds for the treatment of COPD in clinical trials, and discussed their challenges and future development directions. In conclusion, this review combined the latest literature to illustrate the various pathogenesis and interrelationships of COPD in the form of graphs, texts, and tables, and sorted out the functional role and mechanisms of natural polyphenols in treating COPD, with a view to providing new ideas and plans for the in-depth research on COPD and the systemic treatment of COPD with Chinese herbal medicine.
Collapse
Affiliation(s)
- Yang Zhang
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China
| | - Lijuan Wang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Jinyi Zeng
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China
| | - Wen Shen
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China.
| |
Collapse
|
4
|
Vithalkar MP, Pradhan S, Sandra KS, Bharath HB, Nayak Y. Modulating NLRP3 Inflammasomes in Idiopathic Pulmonary Fibrosis: A Comprehensive Review on Flavonoid-Based Interventions. Cell Biochem Biophys 2025:10.1007/s12013-025-01696-4. [PMID: 39966334 DOI: 10.1007/s12013-025-01696-4] [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] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a severe, rapidly advancing disease that drastically diminishes life expectancy. Without treatment, it can progress to lung cancer. The precise etiology of IPF remains unknown, but inflammation and damage to the alveolar epithelium are widely thought to be pivotal in its development. Research has indicated that activating the NLRP3 inflammasome is a crucial mechanism in IPF pathogenesis, as it triggers the release of pro-inflammatory cytokines such as IL-1β, IL-18, and TGF-β. These cytokines contribute to the myofibroblast differentiation and extracellular matrix (ECM) accumulation. Currently, treatment options for IPF are limited. Only two FDA-approved medications, pirfenidone and nintedanib, are available. While these drugs can decelerate disease progression, they come with a range of side effects and do not cure the disease. Additional treatment strategies primarily involve supportive care and therapy. Emerging research has highlighted that numerous flavonoids derived from traditional medicines can inhibit the critical regulators responsible for activating the NLRP3 inflammasome. These flavonoids show promise as potential therapeutic agents for managing IPF, offering a new avenue for treatment that targets the core inflammatory processes of this debilitating condition.
Collapse
Affiliation(s)
- Megh Pravin Vithalkar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India
| | - Shreya Pradhan
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India
| | - K S Sandra
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India
| | - H B Bharath
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, Pin 576104, India.
| |
Collapse
|
5
|
Wang Y, Ma Z, Peng W, Yu Q, Liang W, Cao L, Wang Z. 3,5,6,7,8,3',4'- Heptamethoxyflavonoid inhibits TGF-β1-induced epithelial-mesenchymal transition by regulating oxidative stress and autophagy through MEK/ERK/PI3K/AKT/mTOR signaling pathway. Sci Rep 2025; 15:4567. [PMID: 39915543 PMCID: PMC11802913 DOI: 10.1038/s41598-025-88869-6] [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: 06/12/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial pathological process in the pathogenesis of fibrosis. 3,5,6,7,8,3',4'-hepmethoxyflavone (HMF), the main active ingredient extracted from the Chinese herb Breynia fruticosa (L.) Hook. f., has been shown to have beneficial effects on regulating apoptosis and inhibiting collagen deposition. However, it remains unclear whether and how HMF alleviates transforming growth factor-β1 (TGF-β1)-induced EMT. The objective of this study was to investigate the impact of HMF on TGF-β1-induced EMT in human alveolar Type II epithelial cells (A549) and its underlying mechanism. In vitro culture of TGF-β1-induced EMT in A549 cells revealed that HMF reduced cell viability and migration, inhibited collagen deposition, decreased expression levels of mesenchymal cell markers and fibrosis markers α-SMA, MMP2, TIMP1, β-catenin, and Snail. Meanwhile, the expression level of E-cadherin increased as an epithelial cell marker. Additionally, we discussed the effects of HMF on oxidative stress and autophagy. Various experiments confirmed that HMF regulated the expression levels of Nrf2, keap-1, HO-1, ROS, MDA, SOD, GSH, and played a role in reducing oxidative stress. At the same time, HMF significantly activated autophagy by increasing expressions of Beclin-1 and LC3B as well as enhancing autophagosome content. The addition 3-MA, an autophagy inhibitor attenuated these beneficial effects. Furthermore, HMF significantly inhibited phosphorylation levels of MEK, ERK, PI3K, AKT, and mTOR through various pathways. In conclusion, HMF effectively inhibits TGF-β1-induced EMT in A549 cells by targeting the MEK/ERK/PI3K/AKT/mTOR signaling pathway. Moreover, it exhibits a close correlation with the suppression of oxidative stress and induction of autophagy.
Collapse
Affiliation(s)
- Yiting Wang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Zhiheng Ma
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Weiwen Peng
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Qinglian Yu
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Wenjie Liang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Liu Cao
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Zhuqiang Wang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China.
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China.
| |
Collapse
|
6
|
Shi Z, Zhou M, Zhai J, Sun J, Wang X. Novel therapeutic strategies and drugs for idiopathic pulmonary fibrosis. Arch Pharm (Weinheim) 2024; 357:e2400192. [PMID: 38961537 DOI: 10.1002/ardp.202400192] [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: 03/14/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease of unknown etiology. Currently, drugs used to treat IPF in clinical practice exhibit severe side effects and limitations. To address these issues, this paper discusses the therapeutic effects of preclinical targeted drugs (such as STAT3 and TGF-β/Smad pathway inhibitors, chitinase inhibitors, PI3K and phosphodiesterase inhibitors, etc.) and natural products on IPF. Through a summary of current research progress, it is found that natural products possess multitarget effects, stable therapeutic efficacy, low side effects, and nondrug dependence. Furthermore, we discuss the significant prospects of natural product molecules in combating fibrosis by influencing the immune system, expecting that current analytical data will aid in the development of new drugs or the investigation of active ingredients in natural products for potential IPF treatments in the future.
Collapse
Affiliation(s)
- Zezhou Shi
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Min Zhou
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Jingfang Zhai
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Jie Sun
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Xiaojing Wang
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| |
Collapse
|
7
|
Wang Z, Yang L, Feng Y, Duan B, Zhang H, Tang Y, Zhang C, Yang J. Isoorientin Alleviates DSS-Treated Acute Colitis in Mice by Regulating Intestinal Epithelial P-Glycoprotein (P-gp) Expression. DNA Cell Biol 2024; 43:520-536. [PMID: 39180442 DOI: 10.1089/dna.2024.0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024] Open
Abstract
Isoorientin (ISO) is a naturally occurring flavonoid with diverse functional properties that mitigate the risk of diseases stemming from oxidation, inflammation, and cancer cell proliferation. P-glycoprotein (P-gp) is a vital component of the intestinal epithelium and may play a role in the onset of intestinal inflammatory conditions, such as inflammatory bowel disease (IBD). Recent studies have suggested that short-chain fatty acids (SCFAs) and secondary bile acids (SBAs) produced by the gut microbiota stimulate the increase of P-gp expression, alleviating excessive inflammation and thereby preservation of intestinal homeostasis. ISO has been shown to improve colon health and modulate the gut microbiota. In this study, we aimed to explore whether ISO can modulate the microbes and their metabolites to influence P-gp expression to alleviate IBD. First, the impact of ISO on dextran sulfate sodium (DSS)-treated colitis in mice was investigated. Second, 16S rRNA gene sequencing was conducted. The present study indicated that ISO mitigated the symptoms and pathological damage associated with DSS-treated colitis in mice. Western blot analysis revealed ISO upregulated P-gp in colon tissues, suggesting the critical role of P-gp protein in intestinal epithelial cells. 16S microbial diversity sequencing revealed ISO restored the richness and variety of intestinal microorganisms in colitis-bearing mice and enriched SCFA-producing bacteria, such as Lachnospiraceae_NK4A136_group. The experiments also revealed that the ISO fecal microbiota transplantation (FMT) inoculation of DSS-treated mice had similarly beneficial results. FMT mice showed a reduction in colitis symptoms, which was more pronounced in ISO-FMT than in CON-FMT mice. Meanwhile, ISO-FMT expanded the abundance of beneficial microorganisms, increased the expression of metabolites, such as SCFAs and total SBAs, and significantly upregulated the expression of P-gp protein. In addition, Spearman's correlation analysis demonstrated a positive correlation between the production of SCFAs and SBAs and the expression of P-gp. The present study identified that ISO increases the expression of P-gp in the intestinal epithelium by regulating intestinal microorganisms and their metabolites, which maintains colonic homeostasis, improves the integrity of the colonic epithelium, and alleviates colitis.
Collapse
Affiliation(s)
- Zhenzhen Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Lanzhu Yang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yun Feng
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bensong Duan
- Department of Gastroenterology Shanghai East Hospital, School of Medicine, Endoscopy Center, Tongji University, Shanghai, China
| | - Haibin Zhang
- Department of Gastroenterology Shanghai East Hospital, School of Medicine, Endoscopy Center, Tongji University, Shanghai, China
| | - Yanru Tang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Caihang Zhang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jingya Yang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China
| |
Collapse
|
8
|
Cao M, Huang P, Xu LS, Zhang YH. Analysis of gut microbiota-derived metabolites regulating pituitary neuroendocrine tumors through network pharmacology. Front Pharmacol 2024; 15:1403864. [PMID: 39295931 PMCID: PMC11408289 DOI: 10.3389/fphar.2024.1403864] [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: 05/11/2024] [Accepted: 08/15/2024] [Indexed: 09/21/2024] Open
Abstract
Pituitary neuroendocrine tumors (PitNETs) are a special class of tumors of the central nervous system that are closely related to metabolism, endocrine functions, and immunity. In this study, network pharmacology was used to explore the metabolites and pharmacological mechanisms of PitNET regulation by gut microbiota. The metabolites of the gut microbiota were obtained from the gutMGene database, and the targets related to the metabolites and PitNETs were determined using public databases. A total of 208 metabolites were mined from the gutMGene database; 1,192 metabolite targets were screened from the similarity ensemble approach database; and 2,303 PitNET-related targets were screened from the GeneCards database. From these, 392 overlapping targets were screened between the metabolite and PitNET-related targets, and the intersection between these overlapping and gutMGene database targets (223 targets) were obtained as the core targets (43 targets). Using the protein-protein interaction (PPI) network analysis, Kyoto encyclopedia of genes and genomes (KEGG) signaling pathway and metabolic pathway analysis, CXCL8 was obtained as a hub target, tryptophan metabolism was found to be a key metabolic pathway, and IL-17 signaling was screened as the key KEGG signaling pathway. In addition, molecular docking analysis of the active metabolites and target were performed, and the results showed that baicalin, baicalein, and compound K had good binding activities with CXCL8. We also describe the potential mechanisms for treating PitNETs using the information on the microbiota (Bifidobacterium adolescentis), signaling pathway (IL-17), target (CXCL8), and metabolites (baicalin, baicalein, and compound K); we expect that these will provide a scientific basis for further study.
Collapse
Affiliation(s)
- Min Cao
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Huang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Lun-Shan Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi-Hua Zhang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
9
|
Sharma A, Wairkar S. Flavonoids for treating pulmonary fibrosis: Present status and future prospects. Phytother Res 2024; 38:4406-4423. [PMID: 38986681 DOI: 10.1002/ptr.8285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/08/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with an unknown underlying cause. There is no complete cure for IPF; however, two anti-fibrotic agents (Nintedanib and pirfenidone) are approved by the USFDA to extend the patient's life span. Therefore, alternative therapies supporting the survival of fibrotic patients have been studied in recent literature. The abundance of phenolic compounds, particularly flavonoids, has gathered attention due to their potential health benefits. Various flavonoids, like naringin, quercetin, baicalin, baicalein, puerarin, silymarin, and kaempferol, exhibit anti-inflammatory and anti-oxidant properties, which help decrease lung fibrosis. Various databases, including PubMed, EBSCO, ProQuest, and Scopus, as well as particular websites, such as the World Health Organisation and the National Institutes of Health, were used to conduct a literature search. Several mechanisms of action of flavonoids are reported with the help of in vivo and cell line studies emphasizing their ability to modulate oxidative stress, inflammation, and fibrotic processes in the lungs. They are reported for the restoration of biomarkers like hydroxyproline, cytokines, superoxide dismutase, malondialdehyde and others associated with IPF and for modulating various pathways responsible for the progression of pulmonary fibrosis. Yet, flavonoids have some drawbacks, such as poor solubility, challenging drug loading, stability issues, and scarce bioavailability. Therefore, novel formulations of flavonoids are explored, including liposomes, solid lipid microparticles, polymeric nanoparticles, nanogels, and nanocrystals, to enhance the therapeutic efficacy of flavonoids in pulmonary fibrosis. This review focuses on the role of flavonoids in mitigating idiopathic pulmonary fibrosis, their mode of action and novel formulations.
Collapse
Affiliation(s)
- Anju Sharma
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, India
| |
Collapse
|
10
|
Li Z, Yang Y, Gao F. Monomeric compounds from natural products for the treatment of pulmonary fibrosis: a review. Inflammopharmacology 2024; 32:2203-2217. [PMID: 38724690 DOI: 10.1007/s10787-024-01485-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/23/2024] [Indexed: 08/06/2024]
Abstract
Pulmonary fibrosis (PF) is the end stage of lung injury and chronic lung diseases that results in diminished lung function, respiratory failure, and ultimately mortality. Despite extensive research, the pathogenesis of this disease remains elusive, and effective therapeutic options are currently limited, posing a significant clinical challenge. In addition, research on traditional Chinese medicine and naturopathic medicine is hampered by several complications due to complex composition and lack of reference compounds. Natural product monomers, possessing diverse biological activities and excellent safety profiles, have emerged as potential candidates for preventing and treating PF. The effective anti-PF ingredients identified can be generally divided into flavonoids, saponins, polysaccharides, and alkaloids. Specifically, these monomeric compounds can attenuate inflammatory response, oxidative stress, and other physiopathological processes of the lung through many signaling pathways. They also improve pulmonary factors. Additionally, they ameliorate epithelial-mesenchymal transition (EMT) and fibroblast-myofibroblast transdifferentiation (FMT) by regulating multiple signal amplifiers in the lungs, thereby mitigating PF. This review highlights the significant role of monomer compounds derived from natural products in reducing inflammation, oxidative stress, and inhibiting EMT process. The article provides comprehensive information and serves as a solid foundation for further exploration of new strategies to harness the potential of botanicals in the treatment of PF.
Collapse
Affiliation(s)
- Zhuqing Li
- University of Shanghai for Science and Technology, 516, Jungong Road, Shanghai, 200093, China
| | - Yanyong Yang
- Basic Medical Center for Pulmonary Disease, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, China.
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, People's Republic of China.
| | - Fu Gao
- University of Shanghai for Science and Technology, 516, Jungong Road, Shanghai, 200093, China.
- Basic Medical Center for Pulmonary Disease, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, China.
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, 800, Xiangyin Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
11
|
Xiang G, Yang L, Qin J, Wang S, Zhang Y, Yang S. Revealing the potential bioactive components and mechanism of Qianhua Gout Capsules in the treatment of gouty arthritis through network pharmacology, molecular docking and pharmacodynamic study strategies. Heliyon 2024; 10:e30983. [PMID: 38770346 PMCID: PMC11103544 DOI: 10.1016/j.heliyon.2024.e30983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
Recent clinical studies have confirmed the effectiveness of Qianhua Gout Capsules (QGC) in the treatment of gouty arthritis (GA). However, the specific regulatory targets and mechanisms of action of QGC are still unclear. To address this gap, we utilized network pharmacology, molecular docking, and pharmacodynamic approaches to investigate the bioactive components and associated mechanisms of QGC in the treatment of GA. By employing UPLC-Q Exactive-MS, we identified the compounds present in QGC, with active ingredients defined as those with oral bioavailability ≥30 % and drug similarity ≥0.18. Subsequently, the targets of these active compounds were determined using the TCMSP database, while GA-related targets were identified from DisGeNET, GeneCards, TTD, OMIM, and DrugBank databases. Further analysis including PPI analysis, GO analysis, and KEGG pathway enrichment was conducted on the targets. Validation of the predicted results was performed using a GA rat model, evaluating pathological changes, inflammatory markers, and pathway protein expression. Our results revealed a total of 130 components, 44 active components, 16 potential shared targets, GO-enriched terms, and 47 signaling pathways related to disease targets. Key active ingredients included quercetin, kaempferol, β-sitosterol, luteolin, and wogonin. The PPI analysis highlighted five targets (PPARG, IL-6, MMP-9, IL-1β, CXCL-8) with the highest connectivity, predominantly enriched in the IL-17 signaling pathway. Molecular docking experiments demonstrated strong binding of CXCL8, IL-1β, IL-6, MMP9, and PPARG targets with the top five active compounds. Furthermore, animal experiments confirmed the efficacy of QGC in treating GA in rats, showing reductions in TNF-α, IL-6, and MDA levels, and increases in SOD levels in serum. In synovial tissues, QGC treatment upregulated CXCL8 and PPARG expression, while downregulating IL-1β, MMP9, and IL-6 expression. In conclusion, this study applied a network pharmacology approach to uncover the composition of QGC, predict its pharmacological interactions, and demonstrate its in vivo efficacy, providing insights into the anti-GA mechanisms of QGC. These findings pave the way for future investigations into the therapeutic mechanisms underlying QGC's effectiveness in the treatment of GA.
Collapse
Affiliation(s)
- Gelin Xiang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luyin Yang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Jing Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, China
| | - Yi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, China
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Lao Q, Wang X, Zhu G, Yuan H, Ma T, Wang N. A Chinese classical prescription Maimendong decoction in treatment of pulmonary fibrosis: an overview. Front Pharmacol 2024; 15:1329743. [PMID: 38783956 PMCID: PMC11112100 DOI: 10.3389/fphar.2024.1329743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 04/11/2024] [Indexed: 05/25/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive disease characterized by fibrosis and interstitial pneumonia. It has similar clinical symptoms to "Fei Bi" and "Fei Wei" as described in the traditional Chinese medicine (TCM) classic Jingui Yaolue written by Zhang Zhongjing in the Han Dynasty. This study explored the potential of Maimendong Decoction (MMDD). MMDD consists of Ophiopogon japonicus (L.f) (ophiopogonis), Pinellia ternata (Thunb.) Breit. (pinellia), Panax ginseng C. A. Mey. (ginseng), Glycyrrhiza uralensis Fisch. (glycyrrhiza), Zizi phus jujuba Mill. (jujuba), and Oryza sativa L. (oryza sativa), with the function of nourishing the lung and stomach, and reducing the effect of reverse qi. It has been used clinically for over two thousand years to treat conditions like "Fei Bi" and "Fei Wei". Previous research suggests that MMDD and its individual herbal extracts have anti-fibrotic effects. The main focus of MMDD in treating PF is to reduce inflammatory cytokines, inhibit pro-fibrotic factors and oxidative stress, promote differentiation and homing of bone marrow mesenchymal stem cells, and enhance cell autophagy activity. This review summarized the clinical applications, mechanisms, and pharmacological effects of MMDD in treating PF based on existing clinical applications and experimental research. It also discussed current issues and prospects, aiming to provide a reference for further research on the mechanism of PF, drug development, and clinical trials.
Collapse
Affiliation(s)
- Qiurong Lao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianbin Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangqing Zhu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haochen Yuan
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ting Ma
- College of Rehabilitation Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ning Wang
- Research Department of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Gubareva EA, Golubev AG, Semenov AL, Yurova MN, Kruglov SS, Radetskaya EA. Effects of a Hydrolyzed Lignin Derivative on Bleomycin-Induced Pulmonary Fibrosis in Mice. Bull Exp Biol Med 2024; 177:39-43. [PMID: 38955853 DOI: 10.1007/s10517-024-06127-7] [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: 09/28/2023] [Indexed: 07/04/2024]
Abstract
Female C57BL/J mice with pulmonary fibrosis induced by injections of bleomycin (20 mg/kg intraperitoneally, 8 times for 4 weeks) were treated with a lignin derivative-based composition BP-C3 (80 mg/kg, daily intragastric administrations for 4 weeks). Bleomycin treatment increased the severity of pulmonary fibrosis (Ashcroft score increased from 1.43±0.20 to 4.17±0.48) and the percentage of α-SMA+ tissue (from 15.22±1.01 to 33.12±2.30%) and DNA-synthetizing nuclei (from 1.05±0.14 to 3.38±0.375). After treatment with BP-C3, we observed a tendency to a decrease in Ashcroft score (to 3.40±0.51) and a significant decrease in the percentage of α-SMA+ tissue to 24.30±1.70%; the percentage of DNA-synthetizing nuclei decreased to a lesser extent (to 3.03±0.22%). These results suggest that BP-C3 has a moderate antifibrotic activity.
Collapse
Affiliation(s)
- E A Gubareva
- N. N. Petrov National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, St. Petersburg, Russia.
| | - A G Golubev
- N. N. Petrov National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - A L Semenov
- N. N. Petrov National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - M N Yurova
- N. N. Petrov National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - S S Kruglov
- N. N. Petrov National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - E A Radetskaya
- N. N. Petrov National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| |
Collapse
|
14
|
Cao JL, Li SM, Tang YJ, Hou WS, Wang AQ, Li TZ, Jin CH. Network pharmacology analysis and experimental verification of the antitumor effect and molecular mechanism of isocryptomerin on HepG2 cells. Drug Dev Res 2024; 85:e22165. [PMID: 38400652 DOI: 10.1002/ddr.22165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
Isocryptomerin (ISO) is a flavonoid isolated from the natural medicine Selaginellae Herba, which has various pharmacological activities. This study investigated the antitumor effect and underlying molecular mechanism of ISO on hepatocellular carcinoma (HCC) HepG2 cells. The cell viability assay revealed that ISO has a considerable killing effect on HCC cell lines. The apoptosis assay showed that ISO induced mitochondria-dependent apoptosis through the Bad/cyto-c/cleaved (cle)-caspase-3/cleaved (cle)-PARP pathway. The network pharmacological analysis found 13 key target genes, and epidermal growth factor receptor (EGFR), AKT, mitogen-activated protein kinase (MAPK), and reactive oxygen species (ROS) signaling pathways were strongly associated with ISO against HCC. Further verification of the results showed that ISO induced apoptosis by increasing p-p38 and p-JNK expression and decreasing p-EGFR, p-SRC, p-ERK, and p-STAT3 expression. Furthermore, ISO induced G0/G1 phase arrest by downregulating p-AKT, Cyclin D, and CDK 4 expression and upregulating p21 and p27 expression in HepG2 cells. Moreover, ISO inhibited HepG2 cell migration by decreasing p-GSK-3β, β-catenin, and N-cadherin expression and increasing E-cadherin expression. Additionally, ISO promoted ROS accumulation in HepG2 cells, and ISO-induced apoptosis, arrest cell cycle, and inhibition of migration were reversed by an ROS scavenger, N-acetyl- l-cysteine. Overall, ISO induced cell apoptosis and cell cycle arrest and inhibited cell migration by ROS-mediated EGFR, AKT, and MAPK signaling pathways in HepG2 cells.
Collapse
Affiliation(s)
- Jing-Long Cao
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shu-Mei Li
- Hemodialysis Center, Daqing Oilfield General Hospital, Daqing, China
| | - Yan-Jun Tang
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wen-Shuang Hou
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - An-Qi Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Tian-Zhu Li
- Department of Molecular Biology, College of Basic Medical Science, Chifeng University, Chifeng, China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, China
- National Coarse Cereals Engineering Research Center, Daqing, China
| |
Collapse
|
15
|
Hao B, Yang Z, Liu H, Liu Y, Wang S. Advances in Flavonoid Research: Sources, Biological Activities, and Developmental Prospectives. Curr Issues Mol Biol 2024; 46:2884-2925. [PMID: 38666911 PMCID: PMC11049524 DOI: 10.3390/cimb46040181] [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: 02/02/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
At present, the occurrence of a large number of infectious and non-communicable diseases poses a serious threat to human health as well as to drug development for the treatment of these diseases. One of the most significant challenges is finding new drug candidates that are therapeutically effective and have few or no side effects. In this respect, the active compounds in medicinal plants, especially flavonoids, are potentially useful compounds with a wide range of pharmacological activities. They are naturally present in nature and valuable in the treatment of many infectious and non-communicable diseases. Flavonoids are divided into fourteen categories and are mainly derived from plant extraction, chemical synthesis and structural modification, and biosynthesis. The structural modification of flavonoids is an important way to discover new drugs, but biosynthesis is currently considered the most promising research direction with the potential to revolutionize the new production pipeline in the synthesis of flavonoids. However, relevant problems such as metabolic pathway analyses and cell synthesis protocols for flavonoids need to be addressed on an urgent basis. In the present review, new research techniques for assessing the biological activities of flavonoids and the mechanisms of their biological activities are elucidated and their modes of interaction with other drugs are described. Moreover, novel drug delivery systems, such as nanoparticles, bioparticles, colloidals, etc., are gradually becoming new means of addressing the issues of poor hydrophilicity, lipophilicity, poor chemical stability, and low bioavailability of flavonoids. The present review summarizes the latest research progress on flavonoids, existing problems with their therapeutic efficacy, and how these issues can be solved with the research on flavonoids.
Collapse
Affiliation(s)
| | | | | | | | - Shengyi Wang
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (B.H.); (Z.Y.); (H.L.); (Y.L.)
| |
Collapse
|
16
|
Sharawi ZW, Ibrahim IM, Abd-Alhameed EK, Althagafy HS, Jaber FA, Harakeh S, Hassanein EHM. Baicalin and lung diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1405-1419. [PMID: 37725153 DOI: 10.1007/s00210-023-02704-1] [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: 05/30/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
Studies focusing on natural products have been conducted worldwide, and the results suggest that their natural ingredients effectively treat a wide range of illnesses. Baicalin (BIA) is a glycoside derived from the flavonoid baicalein present in Scutellaria baicalensis of the Lamiaceae family. Interestingly, BIA has been shown to protect the lungs in several animal models used in numerous studies. Therefore, we fully analyzed the data of the studies that focused on BIA's lung protective function against various injuries and included them in this review. Interestingly, BIA exhibits promising effects against acute lung injury, lung fibrosis, pulmonary embolism, and lung remodelling associated with COPD, LPS, and paraquat insecticide. BAI exhibits anticancer activity against lung cancer. Additionally, BIA potently attenuates lung damage associated with infections. BIA primarily exerts its therapeutic effects by suppressing inflammation, oxidative stress immune response, and apoptosis pathways. Nrf2/HO-1, PI3K/Akt, NF-κB, STAT3, MAPKs, TLR4, and NLRP3 are important targets in the pulmonary therapeutic effects of BIA on different lung disease models. Consequently, we recommend using it in future potential clinical applications, its contribution to treatment guidelines, and translating its promising effects to clinical practice in lung diseases.
Collapse
Affiliation(s)
- Zeina W Sharawi
- Biological Sciences Department, Faculty of Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Fatima A Jaber
- Department of Biology, College of Science, University of Jeddah, P.O. Box 80327, Jeddah, 21589, Saudi Arabia
| | - Steve Harakeh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Yousef Abdul Lateef Jameel Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt.
| |
Collapse
|
17
|
Bian B, Ge C, Wu F, Fan Y, Kong J, Li K, Bian H, Miao Q. Wogonin Attenuates Bleomycin-Induced Pulmonary Fibrosis and Oxidative Stress Injury via the MAPK Signaling Pathway. Biol Pharm Bull 2024; 47:2165-2172. [PMID: 39756931 DOI: 10.1248/bpb.b24-00534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Idiopathic pulmonary fibrosis (PF) is an irreversible and chronic inflammatory condition with limited therapeutic options and a high mortality rate. We aimed to determine the possible role and mechanisms of wogonin (WGN) on PF. A rat model of PF was established with intratracheally administrated with bleomycin (BLM), followed by intravenously injecting with WGN and weekly body weight measurements for four weeks. Hematoxylin-eosin (H&E) and Masson's trichrome staining were implemented for histopathological analysis. In addition, the levels of fibrotic proteins and indicators of the mitogen-activated protein kinase (MAPK) pathway were assessed with Western blot. RT-quantitative (q)PCR experiment was conducted to investigate the fibrotic proteins' mRNA expression. Ultimately, the concentrations of glutathione peroxidase (GSH-PX), malonaldehyde (MDA), and superoxide dismutase (SOD) were ascertained with appropriate kits. The results showed that WGN administration significantly reversed BLM-induced body weight reduction, alleviated pathological fibrosis, and reduced the Ashcroft score and the lung wet-to-dry weight ratio. Additionally, WGN suppressed the rise of fibrotic protein levels in BLM-treated rat's lung tissues. Furthermore, WGN attenuated BLM-stimulated oxidative stress, as evidenced by the increased GSH-PX and SOD levels and decreased MDA levels in vivo. Finally, wogonin supplements significantly lowered the extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 MAPK phosphorylation levels in the BLM-treated rat's lung tissues. In conclusion, our study proved that PF induced by BLM administration can be mitigated by WGN treatment via suppressing the MAPK pathway, indicating that WGN is a candidate therapeutic agent for managing PF.
Collapse
Affiliation(s)
- Bo Bian
- Traditional Chinese Medical College, North China University of Science and Technology
| | - Chang Ge
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Fanwu Wu
- Traditional Chinese Medical College, North China University of Science and Technology
| | - Yiling Fan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences
| | | | - Kai Li
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology
| | - Hua Bian
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology
| | - Qing Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences
| |
Collapse
|
18
|
Qin S, Tan P, Xie J, Zhou Y, Zhao J. A systematic review of the research progress of traditional Chinese medicine against pulmonary fibrosis: from a pharmacological perspective. Chin Med 2023; 18:96. [PMID: 37537605 PMCID: PMC10398979 DOI: 10.1186/s13020-023-00797-7] [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: 04/27/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023] Open
Abstract
Pulmonary fibrosis is a chronic progressive interstitial lung disease caused by a variety of etiologies. The disease can eventually lead to irreversible damage to the lung tissue structure, severely affecting respiratory function and posing a serious threat to human health. Currently, glucocorticoids and immunosuppressants are the main drugs used in the clinical treatment of pulmonary fibrosis, but their efficacy is limited and they can cause serious adverse effects. Traditional Chinese medicines have important research value and potential for clinical application in anti-pulmonary fibrosis. In recent years, more and more scientific researches have been conducted on the use of traditional Chinese medicine to improve or reduce pulmonary fibrosis, and some important breakthroughs have been made. This review paper systematically summarized the research progress of pharmacological mechanism of traditional Chinese medicines and their active compounds in improving or reducing pulmonary fibrosis. We conducted a systematic search in several main scientific databases, including PubMed, Web of Science, and Google Scholar, using keywords such as idiopathic pulmonary fibrosis, pulmonary fibrosis, interstitial pneumonia, natural products, herbal medicine, and therapeutic methods. Ultimately, 252 articles were included and systematically evaluated in this analysis. The anti-fibrotic mechanisms of these traditional Chinese medicine studies can be roughly categorized into 5 main aspects, including inhibition of epithelial-mesenchymal transition, anti-inflammatory and antioxidant effects, improvement of extracellular matrix deposition, mediation of apoptosis and autophagy, and inhibition of endoplasmic reticulum stress. The purpose of this article is to provide pharmaceutical researchers with information on the progress of scientific research on improving or reducing Pulmonary fibrosis with traditional Chinese medicine, and to provide reference for further pharmacological research.
Collapse
Affiliation(s)
- Shanbo Qin
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Peng Tan
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China.
| | - Junjie Xie
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Yongfeng Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Junning Zhao
- Key Laboratory of Biological Evaluation of TCM Quality of State Administration of Traditional Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China.
| |
Collapse
|
19
|
Wang MC. Natural plant resource flavonoids as potential therapeutic drugs for pulmonary fibrosis. Heliyon 2023; 9:e19308. [PMID: 37664726 PMCID: PMC10470008 DOI: 10.1016/j.heliyon.2023.e19308] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023] Open
Abstract
Pulmonary fibrosis is an enduring and advancing pulmonary interstitial disease caused by multiple factors that ultimately lead to structural changes in normal lung tissue. Currently, pulmonary fibrosis is a global disease with a high degree of heterogeneity and mortality rate. Nitidine and pirfenidone have been approved for treating pulmonary fibrosis, and the quest for effective therapeutic drugs remains unabated. In recent years, the anti-pulmonary fibrosis properties of natural flavonoids have garnered heightened attention, although further research is needed. In this paper, the resources, structural characteristics, anti-pulmonary fibrosis properties and mechanisms of natural flavonoids were reviewed. We hope to provide potential opportunities for the application of flavonoids in the fight against pulmonary fibrosis.
Collapse
Affiliation(s)
- Meng-Chuan Wang
- Department of Pharmacy, Affiliated Cixi Hospital, Wenzhou Medical University, China
| |
Collapse
|
20
|
Kandeel S, Estfanous RS. Histomorphological and Immunohistochemical Study of Dacomitinib-Induced Ileal Mucositis in Rats with the Possible Protection by Baicalin. J Microsc Ultrastruct 2023; 11:135-144. [PMID: 38025187 PMCID: PMC10679830 DOI: 10.4103/jmau.jmau_115_20] [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: 11/06/2020] [Revised: 09/13/2021] [Accepted: 10/27/2021] [Indexed: 11/10/2022] Open
Abstract
Introduction Gastrointestinal (GIT) mucositis is a common problem associated with chemotherapy. Dacomitinib is a chemotherapeutic drug that treats nonsmall cell lung cancer. It irreversibly binds to the receptors at the ileal epithelial cells, leading to mucosal injury. Baicalin (BA) is a flavonoid with anti-inflammatory, antifibrosis, and antibarrier disruption properties. Aim This work aimed to investigate the possible protective effects of BA on dacomitinib-induced ileal mucositis in rats by histological and immunohistochemical studies. Materials and Methods 60 Wistar rats (8-12 weeks) were used (180-200 g) and divided into 6 groups (10 rats each). Group 1: Control; Group 2 (dacomitinib): Rats received dacomitinib 7.5 mg/kg/day orally; Group 3 (dacomitinib + carboxyl methylcellulose [CMC]): Rats received dacomitinib 7.5 mg/kg/day and 0.5% CMC orally; Group 4 (dacomitinib + BA low dose): Rats received low-dose BA 30 mg/kg/day and 7.5 mg/kg/day dacomitinib orally; Group 5 (dacomitinib + BA mid dose): Rats received mid-dose BA 60 mg/kg/day and 7.5 mg/kg/day dacomitinib orally; Group 6 (dacomitinib + BA high dose): Rats received high-dose BA 100 mg/kg/day and 7.5 mg/kg/day dacomitinib orally. Results Dacomitinib group showed short villi, desquamated epithelium, congested blood vessels, inflammatory cellular infiltrations, dilated lacteals, and wide spaces between the crypts. There is a significant increase in collagen fibers and number of tumor necrosis factor-alpha and proliferating cell nuclear antigen-positive cells. Further, there were lost epithelial cadherin (E-cadherin) and epidermal growth factor receptor immunohistochemical reaction. The previous findings were ameliorated by BA in a dose-dependent manner. Conclusion BA has a protective effect through its anti-inflammatory, antifibrosis, and antibarrier disruption effects. Hence, BA is considered as a promising new drug for the treatment of chemotherapy-associated GIT problems, especially dacomitinib.
Collapse
Affiliation(s)
- Samah Kandeel
- Department Histology and Cell Biology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Remon S. Estfanous
- Department Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
21
|
Ma X, Xia K, Xie J, Yan B, Han X, Li S, Wang Y, Fu T. Treatment of Idiopathic Pulmonary Fibrosis by Inhaled Silybin Dry Powder Prepared via the Nanosuspension Spray Drying Technology. ACS Pharmacol Transl Sci 2023; 6:878-891. [PMID: 37325446 PMCID: PMC10262316 DOI: 10.1021/acsptsci.3c00033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Indexed: 06/17/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a kind of life-threatening interstitial lung disease characterized by progressive dyspnea with accurate pathogenesis unknown. At present, heat shock protein inhibitors are gradually used to treat IPF. Silybin, a heat shock protein C-terminal inhibitor, has high safety and good application prospects. In this work, we have developed a silybin powder able to be used for inhalation administration for the treatment of IPF. Silybin powder was prepared by the spray drying method and identified using cascade impactometry, particle size, scanning electron microscopy (SEM), differential scanning calorimetry (DSC), X-ray diffraction (XRD), and Fourier transform infrared (FT-IR) spectroscopy. A rat model of bleomycin-induced IPF was used to assess the effect of inhaled silybin spray-dried powder. Lung hydroxyproline content, wet weight, histology, inflammatory factor expression, and gene expression were examined. The results showed that inhaled silybin spray-dried powder alleviated inflammation and fibrosis, limited hydroxyproline accumulation in the lungs, modulated gene expression in the development of IPF, and improved postoperative survival. The results of this study suggest that silybin spray-dried powder is an attractive candidate for the treatment of IPF.
Collapse
Affiliation(s)
| | | | - Jianjun Xie
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Baofei Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xingxing Han
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sipan Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yongan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tingming Fu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
22
|
Sun HN, Ren CX, Lee DH, Wang WH, Guo XY, Hao YY, Wang XM, Zhang HN, Xiao WQ, Li N, Cong J, Han YH, Kwon T. PRDX1 negatively regulates bleomycin-induced pulmonary fibrosis via inhibiting the epithelial-mesenchymal transition and lung fibroblast proliferation in vitro and in vivo. Cell Mol Biol Lett 2023; 28:48. [PMID: 37268886 DOI: 10.1186/s11658-023-00460-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/12/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Pulmonary fibrosis is a major category of end-stage changes in lung diseases, characterized by lung epithelial cell damage, proliferation of fibroblasts, and accumulation of extracellular matrix. Peroxiredoxin 1 (PRDX1), a member of the peroxiredoxin protein family, participates in the regulation of the levels of reactive oxygen species in cells and various other physiological activities, as well as the occurrence and development of diseases by functioning as a chaperonin. METHODS Experimental methods including MTT assay, morphological observation of fibrosis, wound healing assay, fluorescence microscopy, flow cytometry, ELISA, western blot, transcriptome sequencing, and histopathological analysis were used in this study. RESULTS PRDX1 knockdown increased ROS levels in lung epithelial cells and promoted epithelial-mesenchymal transition (EMT) through the PI3K/Akt and JNK/Smad signalling pathways. PRDX1 knockout significantly increased TGF-β secretion, ROS production, and cell migration in primary lung fibroblasts. PRDX1 deficiency also increased cell proliferation, cell cycle circulation, and fibrosis progression through the PI3K/Akt and JNK/Smad signalling pathways. BLM treatment induced more severe pulmonary fibrosis in PRDX1-knockout mice, mainly through the PI3K/Akt and JNK/Smad signalling pathways. CONCLUSIONS Our findings strongly suggest that PRDX1 is a key molecule in BLM-induced lung fibrosis progression and acts through modulating EMT and lung fibroblast proliferation; therefore, it may be a therapeutic target for the treatment of BLM-induced lung fibrosis.
Collapse
Affiliation(s)
- Hu-Nan Sun
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China.
| | - Chen-Xi Ren
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Dong Hun Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Wei-Hao Wang
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Xiao-Yu Guo
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Ying-Ying Hao
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Xiao-Ming Wang
- Yabian Academy of Agricultural Science, Longjing, Jilin, 1334000, China
| | - Hui-Na Zhang
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Wan-Qiu Xiao
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Nan Li
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Jie Cong
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Ying-Hao Han
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 351-33 Neongme-Gil, Ibam-Myeon, Jeongeup-Si, Jeonbuk, 56216, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
23
|
Feng T, Duan R, Zheng P, Qiu J, Li Q, Li W. Oxymatrine inhibits TGF‑β1‑mediated mitochondrial apoptotic signaling in alveolar epithelial cells via activation of PI3K/AKT signaling. Exp Ther Med 2023; 25:198. [PMID: 37090069 PMCID: PMC10119625 DOI: 10.3892/etm.2023.11897] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/24/2023] [Indexed: 04/25/2023] Open
Abstract
Although pulmonary fibrosis (PF) causes respiratory failure and death, effective therapies for PF have not been developed. Oxymatrine (OMT), an active ingredient in the Chinese herb Sophora flavescens, exerts antifibrotic effects; however, its effect on PF remains unclear. The present study aimed to determine whether OMT decreases transforming growth factor-β1 (TGF-β1)-induced PF in human lung cancer A549 cells by inhibiting apoptosis and targeting the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway. To construct a PF cell model, A549 cells were stimulated with TGF-β1. The experimental groups were as follows: control (untreated cells grown in complete medium), TGF-β1 (cells treated with 5 ng/ml TGF-β1), OMT (cells treated with 5 ng/ml TGF-β1 and 0.25, 0.50, or 1.00 mg/ml OMT), and OMT + LY294002 (cells treated with 5 ng/ml TGF-β1, 1.0 mg/ml OMT. and 25 µmol/l LY294002). The effects of OMT on cell morphology (via electron microscopy), apoptosis (via Annexin V-PI staining), mitochondrial apoptosis signaling [using JC-1 method to analyze mitochondrial membrane potential (MMP)], and Bcl-2, as well as Bax expression (via western blotting and reverse transcription-quantitative polymerase chain reaction), were analyzed. OMT significantly protected cells against TGF-β1-induced PF by inhibiting apoptosis. The specific manifestations were cell injury, as evidenced by morphological changes and decreased MMP. Following OMT treatment, the expression of the pro-apoptotic protein Bax increased, whereas that of the anti-apoptotic protein Bcl-2 decreased. The PI3K/AKT-specific inhibitor LY294002 significantly inhibited the ameliorative effects of OMT on TGF-β1-induced apoptosis. Collectively, OMT attenuated TGF-β1-mediated mitochondrial apoptosis of alveolar epithelial cells by activating the PI3K/AKT signaling pathway. Therefore, OMT may be a promising drug for PF treatment.
Collapse
Affiliation(s)
- Tong Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Key Laboratory of Geriatrics Respiratory Disease Education Department of Sichuan, Chengdu, Sichuan 610500, P.R. China
| | - Ran Duan
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Department of Cardiology, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Pengcheng Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Jing Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Qingyuan Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Key Laboratory of Geriatrics Respiratory Disease Education Department of Sichuan, Chengdu, Sichuan 610500, P.R. China
| | - Wancheng Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
- Key Laboratory of Geriatrics Respiratory Disease Education Department of Sichuan, Chengdu, Sichuan 610500, P.R. China
- Correspondence to: Professor Wancheng Li, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Avenue, Xindu, Chengdu, Sichuan 610599, P.R. China
| |
Collapse
|
24
|
Zhang Y, Liu F, Jia Q, Zheng L, Tang Q, Sai L, Zhang W, Du Z, Peng C, Bo C, Zhang F. Baicalin alleviates silica-induced lung inflammation and fibrosis by inhibiting TLR4/NF-?B pathway in rats. Physiol Res 2023; 72:221-233. [PMID: 37159856 PMCID: PMC10226396 DOI: 10.33549/physiolres.934978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/22/2022] [Indexed: 03/24/2024] Open
Abstract
Silicosis is an occupational lung disease caused by inhaling silica dust. The disease is characterized by early lung inflammation and late irreversible pulmonary fibrosis. Here we report the effect of Baicalin, a main flavonoid compound from the roots of Chinese herbal medicine Huang Qin on silicosis in a rat model. Results showed Baicalin (50 or 100 mg/kg/day) can mitigate the silica-induced lung inflammation and reduce the harm of alveolar structure and the blue region of collagen fibers in rat lung at 28 days after administration. At the same time, Baicalin also diminished the level of interleukin-1beta (IL-1beta, interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-alpha) and transforming growth factor-beta1 (TGF-beta1) in lung tissues. The protein expression of collagen I (Col-1), alpha-smooth muscle actin (alpha-SMA) and vimentin were down-regulated while E-cadherin (E-cad) was increased in Baicalin-treated rats. In addition, the Toll Like Receptor 4 (TLR4)/ nuclear factor kappaB (NF-kappaB) pathway was enabled at 28 days after silica infusion, and the treatment of Baicalin diminished the expression of TLR4 and NF-?B in the lungs of rat with silicosis. These results suggested that Baicalin inhibited the pulmonary inflammatory and fibrosis in a rat model of silicosis, which could be attributed to inhibition of the TLR4/NF-kappaB pathway.
Collapse
Affiliation(s)
- Y Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China. ,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Tekeli MY, Eraslan G, Bayram LÇ, Aslan C, Çalımlı S. The protective effects of baicalin and chrysin against emamectin benzoate-induced toxicity in Wistar albino rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:53997-54021. [PMID: 36869176 DOI: 10.1007/s11356-023-26110-5] [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: 07/14/2022] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
The aim of this study was to investigate the effects of baicalin, chrysin and their combinations against emamectin benzoate-induced toxicity in rats. For this purpose, sixty four rats were divided into evenly 8 groups with 6-8-week-old male Wistar albino rats, weighing 180-250 g, in each group. While the first group was kept as a control (corn oil), the remaining 7 groups were administered with emamectin benzoate (10 mg/kg bw), baicalin (50 mg/kg bw) and chrysin (50 mg/kg bw) alone or together for 28 days. Oxidative stress parameters, serum biochemical parameters and blood/tissue (liver, kidney, brain, testis and heart) and tissue histopathology were investigated. Compared to the control group, the emamectin benzoate-intoxicated rats had significantly higher tissue/plasma concentrations of nitric oxide (NO) and malondialdehyde (MDA), as well as lower tissue glutathione (GSH) concentrations and antioxidant enzyme activity (glutathione peroxidase/GSH-Px, glutathione reductase/GR, glutathione-S-transferase/GST, superoxide dismutase/SOD, catalase/CAT). Biochemical analysis showed that emamectin benzoate administration significantly increased serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP) and lactate dehydrogenase (LDH) activities, as well as triglyceride, cholesterol, creatinine, uric acid and urea levels, and decreased serum total protein and albumin levels. The histopathological examination of the liver, kidney, brain, heart and testis tissues of the emamectin benzoate-intoxicated rats demonstrated necrotic changes. Baicalin and/or chrysin reversed the biochemical and histopathological alterations induced by emamectin benzoate on these tested organs. Therefore, baicalin and chrysin (alone or in combination) could offer protection against emamectin benzoate-induced toxicity.
Collapse
Affiliation(s)
- Muhammet Yasin Tekeli
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Gökhan Eraslan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey.
| | - Latife Çakır Bayram
- Department of Pathology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Coşkun Aslan
- Derinkuyu Emineana and Yaşar Ertaş Agriculture and Livestock Vocational School, Nevşehir Hacı Bektaş Veli University, Nevşehir, Turkey
| | - Sinem Çalımlı
- Department of Veterinary Pharmacology and Toxicology, Institute of Health Science, Erciyes University, Kayseri, Turkey
| |
Collapse
|
26
|
Song S, Ding L, Liu G, Chen T, Zhao M, Li X, Li M, Qi H, Chen J, Wang Z, Wang Y, Ma J, Wang Q, Li X, Wang Z. The protective effects of baicalin for respiratory diseases: an update and future perspectives. Front Pharmacol 2023; 14:1129817. [PMID: 37007037 PMCID: PMC10060540 DOI: 10.3389/fphar.2023.1129817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/17/2023] Open
Abstract
Background: Respiratory diseases are common and frequent diseases. Due to the high pathogenicity and side effects of respiratory diseases, the discovery of new strategies for drug treatment is a hot area of research. Scutellaria baicalensis Georgi (SBG) has been used as a medicinal herb in China for over 2000 years. Baicalin (BA) is a flavonoid active ingredient extracted from SBG that BA has been found to exert various pharmacological effects against respiratory diseases. However, there is no comprehensive review of the mechanism of the effects of BA in treating respiratory diseases. This review aims to summarize the current pharmacokinetics of BA, baicalin-loaded nano-delivery system, and its molecular mechanisms and therapeutical effects for treating respiratory diseases.Method: This review reviewed databases such as PubMed, NCBI, and Web of Science from their inception to 13 December 2022, in which literature was related to “baicalin”, “Scutellaria baicalensis Georgi”, “COVID-19”, “acute lung injury”, “pulmonary arterial hypertension”, “asthma”, “chronic obstructive pulmonary disease”, “pulmonary fibrosis”, “lung cancer”, “pharmacokinetics”, “liposomes”, “nano-emulsions”, “micelles”, “phospholipid complexes”, “solid dispersions”, “inclusion complexes”, and other terms.Result: The pharmacokinetics of BA involves mainly gastrointestinal hydrolysis, the enteroglycoside cycle, multiple metabolic pathways, and excretion in bile and urine. Due to the poor bioavailability and solubility of BA, liposomes, nano-emulsions, micelles, phospholipid complexes, solid dispersions, and inclusion complexes of BA have been developed to improve its bioavailability, lung targeting, and solubility. BA exerts potent effects mainly by mediating upstream oxidative stress, inflammation, apoptosis, and immune response pathways. It regulates are the NF-κB, PI3K/AKT, TGF-β/Smad, Nrf2/HO-1, and ERK/GSK3β pathways.Conclusion: This review presents comprehensive information on BA about pharmacokinetics, baicalin-loaded nano-delivery system, and its therapeutic effects and potential pharmacological mechanisms in respiratory diseases. The available studies suggest that BA has excellent possible treatment of respiratory diseases and is worthy of further investigation and development.
Collapse
Affiliation(s)
- Siyu Song
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Lu Ding
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guangwen Liu
- GCP Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Tian Chen
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Meiru Zhao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xueyan Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Min Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongyu Qi
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jinjin Chen
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ziyuan Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Ying Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Ma
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Qi Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- *Correspondence: Xiangyan Li, ; Zeyu Wang,
| | - Zeyu Wang
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- *Correspondence: Xiangyan Li, ; Zeyu Wang,
| |
Collapse
|
27
|
Wang D, Li Y. Pharmacological effects of baicalin in lung diseases. Front Pharmacol 2023; 14:1188202. [PMID: 37168996 PMCID: PMC10164968 DOI: 10.3389/fphar.2023.1188202] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
The flavonoids baicalin and baicalein were discovered in the root of Scutellaria baicalensis Georgi and are primarily used in traditional Chinese medicine, herbal supplements and healthcare. Recently, accumulated investigations have demonstrated the therapeutic benefits of baicalin in treating various lung diseases due to its antioxidant, anti-inflammatory, immunomodulatory, antiapoptotic, anticancer, and antiviral effects. In this review, the PubMed database and ClinicalTrials website were searched with the search string "baicalin" and "lung" for articles published between September 1970 and March 2023. We summarized the therapeutic role that baicalin plays in a variety of lung diseases, such as chronic obstructive pulmonary disease, asthma, pulmonary fibrosis, pulmonary hypertension, pulmonary infections, acute lung injury/acute respiratory distress syndrome, and lung cancer. We also discussed the underlying mechanisms of baicalin targeting in these lung diseases.
Collapse
Affiliation(s)
- Duoning Wang
- Chengdu Hi-tech Nanxili Jiuzheng Clinic, Chengdu, Sichuan, China
| | - Yi Li
- Chengdu Hi-tech Nanxili Jiuzheng Clinic, Chengdu, Sichuan, China
- *Correspondence: Yi Li, /
| |
Collapse
|
28
|
Huang Y, Wang A, Jin S, Liu F, Xu F. Activation of the NLRP3 inflammasome by HMGB1 through inhibition of the Nrf2/HO-1 pathway promotes bleomycin-induced pulmonary fibrosis after acute lung injury in rats. Allergol Immunopathol (Madr) 2023; 51:56-67. [PMID: 37169561 DOI: 10.15586/aei.v51i3.668] [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: 05/12/2022] [Accepted: 09/23/2022] [Indexed: 05/13/2023]
Abstract
OBJECTIVE Acute lung injury (ALI) is a common complication of critical diseases with high morbidity and mortality. This study explored the regulatory role and mechanism of high mobility histone box 1 protein (HMGB1) on pulmonary fibrosis (PF) after ALI in rats through nucleotide oligomerization domain-like receptor protein-3 (NLRP3) inflammasome. METHODS PF rat models after ALI were established by induction of bleomycin. Degree of fibrosis was assessed by Masson staining and Ashcroft scoring. Hydroxyproline (Hyp) contents in lung tissues and rat lung tissue morphology were detected by enzyme-linked-immunosorbent serologic assay (ELISA) and hematoxylin and eosin staining. The levels of NLRP3, major proteins of NLRP3 inflammasome (NLRP3/ASC/caspase-1), and downstream inflammatory cytokines interleukin (IL)-1 and IL-18 were determined using immunohistochemistry, Western blotting analysis, and ELISA. The nuclear/cytoplasmic nuclear factor erythroid 2-related factor 2 (Nrf2) levels and HO-1 levels were measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blotting analysis. Rats was injected with lentivirus carrying short hairpin (sh)-HMGB1 and zinc protoporphyria (ZNPP) (HO-1 inhibitor) to assess the effects of HMGB1 and HO-1 on PF and NLRP3 inflammasome activation. RESULTS Bleomycin induced PF after ALI in rats, manifested as patchy fibrosis, atelectasis, and excessive expansion, and increased Aschcroft score and Hyp content. Bleomycin treatment enhanced levels of NLRP3, ASC, caspase-1, IL-1, and IL-18 in rat lung tissues, which promoted activation of NLRP3 inflammasome. HMGB1 was up-regulated in bleomycin-induced rats. HMGB1 knockdown partially reversed NLRP3 inflammasome activation and PF progression. HMGB1 knockdown promoted Nrf2 nuclear translocation and up-regulated HO-1. Suppression of HO-1 partially reversed inhibition of HMGB1 knockdown on NLRP3 inflammasome activation and PF. CONCLUSION HMGB1 can activate NLRP3 inflammasomes and promote PF by inhibiting the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Ying Huang
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan, Hubei Province, China
| | - Aili Wang
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan, Hubei Province, China
| | - Sheng Jin
- Nephrology Department of Integrated Traditional Chinese and Western Medicine, Hubei No. 3 People's Hospital of Jianghan University, Wuhan 430033, Hubei Province, China
| | - Fang Liu
- Department of Respiratory Medicine, Hubei No. 3 People's Hospital of Jianghan University, Wuhan 430033, Hubei Province, China;
| | - Fang Xu
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan, Hubei Province, China;
| |
Collapse
|
29
|
Calycosin Ameliorates Bleomycin-Induced Pulmonary Fibrosis via Suppressing Oxidative Stress, Apoptosis, and Enhancing Autophagy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9969729. [PMID: 36267093 PMCID: PMC9578840 DOI: 10.1155/2022/9969729] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/05/2022]
Abstract
Calycosin (CA) is a flavonoid extracted from the root of Astragalus membranaceus and has antioxidant, anti-inflammation, and antiapoptosis properties. The objective of this study was to investigate the efficacy of CA in protecting against pulmonary fibrosis. CA (14 mg/kg) and SB216763 (20 mg/kg) were administrated to bleomycin-induced pulmonary fibrosis mice for 3 weeks. The results concluded that CA alleviated the inflammation and collagen deposition in pulmonary fibrosis. In addition, CA reduced MDA level, enhanced SOD and TAC activities, and increased the activity of the Nrf2/HO-1 pathway. CA also regulated the expressions of apoptosis-related proteins. Moreover, CA enhanced autophagy via upregulating LC3, beclin1, PINK1, and reducing p62. CA also increased expression of LAMP1 and TFEB, and inhibited the release of lysosome enzymes from ruptured lysosomes. These results provide new evidence that CA protects against pulmonary fibrosis through inhibiting oxidative stress and apoptosis. In addition, autophagy abnormality and lysosome dysfunction are restored by CA.
Collapse
|
30
|
Huang TT, Chen CM, Chen LG, Lan YW, Huang TH, Choo KB, Chong KY. 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucoside ameliorates bleomycin-induced pulmonary fibrosis via regulating pro-fibrotic signaling pathways. Front Pharmacol 2022; 13:997100. [PMID: 36267283 PMCID: PMC9577370 DOI: 10.3389/fphar.2022.997100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/12/2022] [Indexed: 12/01/2022] Open
Abstract
2,3,5,4′-Tetrahydroxystilbene-2-O-β-D-Glucoside (THSG) is the main active ingredient extracted from Polygonum multiflorum Thunb. (PMT), which has been reported to possess extensive pharmacological properties. Nevertheless, the exact role of THSG in pulmonary fibrosis has not been demonstrated yet. The main purpose of this study was to investigate the protective effect of THSG against bleomycin (BLM)-induced lung fibrosis in a murine model, and explore the underlying mechanisms of THSG in transforming growth factor-beta 1 (TGF-β1)-induced fibrogenesis using MRC-5 human lung fibroblast cells. We found that THSG significantly attenuated lung injury by reducing fibrosis and extracellular matrix deposition. THSG treatment significantly downregulated the expression levels of TGF-β1, fibronectin, α-SMA, CTGF, and TGFBR2, however, upregulated the expression levels of antioxidants (SOD-1 and catalase) and LC3B in the lungs of BLM-treated mice. THSG treatment decreased the expression levels of fibronectin, α-SMA, and CTGF in TGF-β1-stimulated MRC-5 cells. Conversely, THSG increased the expression levels of SOD-1 and catalase. Furthermore, treatment of THSG profoundly reduced the TGF-β1-induced generation of reactive oxygen species (ROS). In addition, THSG restored TGF-β1-induced impaired autophagy, accompany by increasing the protein levels of LC3B-II and Beclin 1. Mechanism study indicated that THSG significantly reduced TGF-β1-induced increase of TGFBR2 expression and phosphorylation of Smad2/3, Akt, mTOR, and ERK1/2 in MRC-5 cells. These findings suggest that THSG may be considered as an anti-fibrotic drug for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Tsung-Teng Huang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Division of Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
- The iEGG and Animal Biotechnology Center and the Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Lih-Geeng Chen
- Department of Microbiology, Immunology and Biopharmaceuticals, National Chiayi University, Chiayi, Taiwan
| | - Ying-Wei Lan
- Division of Pulmonary Biology, The Perinatal Institute of Cincinnati Children’s Research Foundation, Cincinnati, OH, United States
| | - Tse-Hung Huang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| | - Kong Bung Choo
- Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, Selangor, Malaysia
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Division of Biotechnology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
- Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, Selangor, Malaysia
- Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- *Correspondence: Kowit-Yu Chong,
| |
Collapse
|
31
|
Wu X, Gou H, Zhou O, Qiu H, Liu H, Fu Z, Chen L. Human umbilical cord mesenchymal stem cells combined with pirfenidone upregulates the expression of RGS2 in the pulmonary fibrosis in mice. Respir Res 2022; 23:270. [PMID: 36182915 PMCID: PMC9526322 DOI: 10.1186/s12931-022-02192-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
Objective The therapeutic effect of umbilical cord-derived mesenchymal stem cells (hUC-MSCs) in combination with pirfenidone (PFD) on pulmonary fibrosis in mice and its possible mechanism were investigated. Methods C57BL/6 mice were randomly divided into six groups: control group, model group, P10 group, P30 group, P100 group, and P300 group. Modeled by tracheal intubation with 3 mg/kg bleomycin drip, each dose of PFD was administered daily by gavage from day 7 onwards. The mice were observed continuously for 21 days and survival was recorded. Lung tissues were collected on day 21, and hematoxylin–eosin (HE) and Masson staining were performed to assess morphological changes and collagen deposition in the lungs. Collagen content was measured by the Sircol method, and fibrosis marker levels were detected by PCR and Western blot. Another batch of C57BL/6 mice was then randomly divided into five groups: hUC-MSC control group, model group, P100 group, hUC-MSC treatment group, and hUC-MSCs + P30 group. On day 7, 5 × 105 hUC-MSCs were injected into the tail vein, the mice were administered PFD gavage daily from day 7 onwards, and their survival was recorded. Lung tissues were collected on day 21 to detect pathological changes, the collagen content, and the expression of regulator of G protein signaling 2 (RGS2). Pulmonary myofibroblasts (MFBs) were divided into an MFB group and an MFB + hUC-MSCs group; different doses of PFD were administered to each group, and the levels of RGS2, intracellular Ca2+, and fibrosis markers were recorded for each group. Results Compared with other PFD group doses, the P100 group had significantly improved mouse survival and lung pathology and significantly reduced collagen and fibrosis marker levels (p < 0.05). The hUC-MSCs + P30 group had significantly improved mouse survival and lung pathology, significantly reduced collagen content and fibrosis marker levels (p < 0.05), and the efficacy was better than that of the P100 and hUC-MSCs groups (p < 0.05). RGS2 expression was significantly higher in the MSCs + P30 group compared with the P100 and hUC-MSCs groups (p < 0.05). PFD increased RGS2 expression in MFBs (p < 0.05) in a dose-dependent manner. Compared with PFD and hUC-MSCs treatment alone, combination of hUC-MSCs and PFD increased RGS2 protein levels, significantly decreased intracellular Ca2+ concentration, and significantly reduced fibrosis markers. Conclusion The findings suggest that hUC-MSCs combined with low-dose PFD have a therapeutic effect better than that of the two treatments used separately. Its effect on attenuating bleomycin-induced pulmonary fibrosis in mice is related to the increase of RGS2. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02192-6.
Collapse
Affiliation(s)
- Xian Wu
- Division of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, Sichuan, China.,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Hao Gou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Ou Zhou
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Huijun Qiu
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Hanmin Liu
- Division of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, Sichuan, China.,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Zhou Fu
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China. .,Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China. .,Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, 400015, China.
| | - Lina Chen
- Division of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China. .,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, Sichuan, China. .,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, 610065, Sichuan, China.
| |
Collapse
|
32
|
Contribution of Adiponectin/Carnitine Palmityl Transferase 1A-Mediated Fatty Acid Metabolism during the Development of Idiopathic Pulmonary Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5265616. [PMID: 36035217 PMCID: PMC9402305 DOI: 10.1155/2022/5265616] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/14/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease that leads rapidly to death. The present study is aimed at discovering the in-depth pathogenesis of IPF, exploring the role of adiponectin/carnitine palmityl transferase 1A- (APN/CPT1A-) mediated fatty acid metabolism during the development of IPF, and excavating its potential mechanism. Here, THP-1 cells were differentiated into M0 macrophages, followed by polarization to M1 macrophages upon hypoxia. Subsequently, lung fibroblast HFL-1 cells were stimulated by M1 macrophages to simulate hypoxia-related IPF condition in vitro. It was discovered that the stimulation of M1 macrophages promoted fibroblast proliferation and fibrosis formation in vitro, accompanied with a disorder of the APN/CPT1A pathway, an overproduction of lipid peroxides, and a low level of autophagy in HFL-1 cells. Thereafter, APN treatment or CPT1A overexpression greatly suppressed above lipid peroxide accumulation, fibroblast proliferation, and fibrosis but activated autophagy in vitro. Furthermore, an in vivo IPF rat model was established by injection of bleomycin (BLM). Consistently, CPT1A overexpression exerted a protective role against pulmonary fibrosis in vivo; however, the antifibrosis property of CPT1A was partly abolished by 3-methyladenine (an autophagy inhibitor). In summary, APN/CPT1A-mediated fatty acid metabolism exerted its protective role in IPF partly through activating autophagy, shedding a new prospective for the treatment of IPF.
Collapse
|
33
|
Scutellaria baicalensis and its constituents baicalin and baicalein as antidotes or protective agents against chemical toxicities: a comprehensive review. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:1297-1329. [PMID: 35676380 DOI: 10.1007/s00210-022-02258-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/21/2022] [Indexed: 10/18/2022]
Abstract
Scutellaria baicalensis (SB), also known as the Chinese skullcap, has a long history of being used in Chinese medicine to treat a variety of conditions ranging from microbial infections to metabolic syndrome and malignancies. Numerous studies have reported that treatment with total SB extract or two main flavonoids found in its root and leaves, baicalin (BA) and baicalein (BE), can prevent or alleviate the detrimental toxic effects of exposure to various chemical compounds. It has been shown that BA and BE are generally behind the protective effects of SB against toxicants. This paper aimed to review the protective and therapeutic effects of SB and its main components BA and BE against chemical compounds that can cause intoxication after acute or chronic exposure and seriously affect different vital organs including the brain, heart, liver, and kidneys. In this review paper, we had a look into a total of 221 in vitro and in vivo studies from 1995 to 2021 from the scientific databases PubMed, Scopus, and Web of Science which reported protective or therapeutic effects of BA, BE, or SB against drugs and chemicals that one might be exposed to on a professional or accidental basis and compounds that are primarily used to simulate disease models. In conclusion, the protective effects of SB and its flavonoids can be mainly attributed to increase in antioxidants enzymes, inhibition of lipid peroxidation, reduction of inflammatory cytokines, and suppression of apoptosis pathway.
Collapse
|
34
|
Elhady SS, Goda MS, Mehanna ET, Elfaky MA, Koshak AE, Noor AO, Bogari HA, Malatani RT, Abdelhameed RFA, Wahba AS. Meleagrin Isolated from the Red Sea Fungus Penicillium chrysogenum Protects against Bleomycin-Induced Pulmonary Fibrosis in Mice. Biomedicines 2022; 10:biomedicines10051164. [PMID: 35625905 PMCID: PMC9138525 DOI: 10.3390/biomedicines10051164] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 12/18/2022] Open
Abstract
The Red Sea marine fungus Penicillium chrysogenum (Family: Ascomycota) comprises a panel of chemically diverse natural metabolites. A meleagrin alkaloid was isolated from deep-sediment-derived P. chrysogenum Strain S003 and has been reported to exert antibacterial and cytotoxic activities. The present study aimed to explore the therapeutic potential of meleagrin on pulmonary fibrosis. Lung fibrosis was induced in mice by a single intratracheal instillation of 2.5 mg/kg bleomycin. Mice were given 5 mg/kg meleagrin daily either for 3 weeks after bleomycin administration in the treatment group or 2 weeks before and 3 weeks after bleomycin administration in the protection group. Bleomycin triggered excessive ROS production, inflammatory infiltration, collagen overproduction and fibrosis. Bleomycin-induced pulmonary fibrosis was attenuated by meleagrin. Meleagrin was noted to restore the oxidant–antioxidant balance, as evidenced by lower MDA contents and higher levels of SOD and catalase activities and GSH content compared to the bleomycin group. Meleagrin also activated the Nrf2/HO-1 antioxidant signaling pathway and inhibited TLR4 and NF-κB gene expression, with a subsequent decreased release of pro-inflammatory cytokines (TNF-α, IL-6 and IFN-γ). Additionally, meleagrin inhibited bleomycin-induced apoptosis by abating the activities of pro-apoptotic proteins Bax and caspase-3 while elevating Bcl2. Furthermore, it suppressed the gene expression of α-SMA, TGF-β1, Smad-2, type I collagen and MMP-9, with a concomitant decrease in the protein levels of TGF-β1, α-SMA, phosphorylated Smad-2, MMP-9, elastin and fibronectin. This study revealed that meleagrin’s protective effects against bleomycin-induced pulmonary fibrosis are attributed to its antioxidant, anti-inflammatory, anti-apoptotic and antifibrotic properties. Notably, the use of meleagrin as a protective agent against bleomycin-induced lung fibrosis was more efficient than its use as a treatment agent.
Collapse
Affiliation(s)
- Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.A.E.); (A.E.K.)
- Correspondence: (S.S.E.); (E.T.M.); Tel.: +966-544512552 (S.S.E.)
| | - Marwa S. Goda
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| | - Eman T. Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
- Correspondence: (S.S.E.); (E.T.M.); Tel.: +966-544512552 (S.S.E.)
| | - Mahmoud A. Elfaky
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.A.E.); (A.E.K.)
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulrahman E. Koshak
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.A.E.); (A.E.K.)
| | - Ahmad O. Noor
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.O.N.); (H.A.B.); (R.T.M.)
| | - Hanin A. Bogari
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.O.N.); (H.A.B.); (R.T.M.)
| | - Rania T. Malatani
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.O.N.); (H.A.B.); (R.T.M.)
| | - Reda F. A. Abdelhameed
- Department of Pharmacognosy, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt;
| | - Alaa S. Wahba
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| |
Collapse
|
35
|
Xu H, Yang J, Tu M, Weng J, Xie M, Zhou Z, Zhou P, Wang L, Chen C, Wang Z. Vincristine Promotes Transdifferentiation of Fibroblasts Into Myofibroblasts via P38 and ERK Signal Pathways. Front Pharmacol 2022; 13:901000. [PMID: 35614948 PMCID: PMC9124770 DOI: 10.3389/fphar.2022.901000] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Vincristine (VCR) is used in the clinic as an anti-tumor drug. VCR can cause pulmonary fibrosis (PF), leading to respiratory failure. The transformation of fibroblasts into myofibroblasts may play a key role in PF. The present study attempted to reveal the molecular mechanism of VCR-induced PF and the possible involvement of the mitogen-activated protein kinase (MAPK) signaling pathway.Methods: Human embryonic lung fibroblasts (HELFs) were treated with different concentrations of VCR. Inhibitors of extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 MAPK were added to HELFs. Cell proliferation state was assessed using cell counting kit-8 and by directly counting the number of cells. The expressions of vimentin and α-smooth muscle actin (α-SMA) were investigated using western blot and immunofluorescence analyses. Activation of ERK and P38 was estimated by the expression of phosphorylated p38 MAPK (p-p38), p38 MAPK, phosphorylated ERK1/2 (p-ERK1/2) and ERK1/2 using western blot analysis. Enzyme-linked immunosorbent assay was used to estimate the level of collagen I in cell culture supernatants.Results: Results showed that VCR promoted cellular proliferation, secretion of collagen I and the expression of vimentin and α-SMA. High expression of p-p38 and p-ERK1/2 was associated with the activation of the MAPK signaling pathway. MAPK inhibitors SB203580 and PD98059 suppressed the expression of the above proteins.Conclusion: Our study revealed that VCR could promote the differentiation of fibroblasts into myofibroblasts by regulating the MAPK signal pathway, which may be a promising way to treat VCR-induced PF.
Collapse
Affiliation(s)
- Hui Xu
- Department of General Practice, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingwen Yang
- Department of Geriatric Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengyun Tu
- Department of Clinical Laboratory, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Weng
- Department of General Practice, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengying Xie
- Department of General Practice, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiliang Zhou
- Department of General Practice, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peisen Zhou
- Department of Emergency Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liang Wang
- Department of Public Health, Robbins College of Health and Human Sciences, Baylor University, Waco, TX, United States
| | - Chan Chen
- Department of Geriatric Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Bioscaffold Transplantation and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zhiyi Wang, ; Chan Chen,
| | - Zhiyi Wang
- Department of General Practice, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zhiyi Wang, ; Chan Chen,
| |
Collapse
|
36
|
Ding L, Li Y, Yang Y, Song S, Qi H, Wang J, Wang Z, Zhao J, Zhang W, Zhao L, Zhao D, Li X, Wang Z. Wenfei Buqi Tongluo Formula Against Bleomycin-Induced Pulmonary Fibrosis by Inhibiting TGF-β/Smad3 Pathway. Front Pharmacol 2022; 12:762998. [PMID: 35126110 PMCID: PMC8814462 DOI: 10.3389/fphar.2021.762998] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/14/2021] [Indexed: 01/06/2023] Open
Abstract
Pulmonary fibrosis (PF) is the end stage of various chronic and progressive interstitial lung diseases. TGF-β, a profibrotic cytokine, can promote epithelial–mesenchymal transition (EMT), extracellular matrix (ECM) accumulation, and fibroblast proliferation, which contribute to progressive lung remodeling in PF. The Wenfei Buqi Tongluo (WBT) formula has been certified to be effective in the prevention and treatment of PF in clinical practice and has inhibitory effects on EMT, inflammation, and profibrotic factors. However, the pharmacological mechanisms of WBT against PF need to be further explored. In this study, we first analyzed the chemical components of the WBT formula using the UHPLC/Q-TOF-MS analysis. The potential targets of the identified compounds from WBT were predicted by the network pharmacology, which was confirmed by in vivo and in vitro study. After screening by the PubChem database, we first identified the 36 compounds of WBT and predicted the TGF-β signaling pathway, with ECM degradation as potential mechanism of WBT against PF by the network pharmacology. Furthermore, WBT treatment inhibited the levels of TGF-β and Smad3 phosphorylation and subsequently alleviated EMT and ECM accumulation in the bleomycin-induced mouse model and TGF-β1–induced cell model. These findings indicate that WBT can block the progressive process of PF by inhibiting EMT and promoting ECM degradation via the TGF-β/Smad3 pathway. This study may provide new insights into the molecular mechanism of WBT for the prevention and treatment of PF in the clinical application.
Collapse
Affiliation(s)
- Lu Ding
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yaxin Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yingying Yang
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Siyu Song
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongyu Qi
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Wang
- Department of Respiratory, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Ziyuan Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jiachao Zhao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Wei Zhang
- Department of Scientific Research, Changchun University of Chinese Medicine, Changchun, China
| | - Linhua Zhao
- Molecular Biology Laboratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zeyu Wang
- Department of Scientific Research, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
37
|
Bai S, Peng X, Wu C, Cai T, Liu J, Shu G. Effects of dietary inclusion of Radix Bupleuri extract on the growth performance, and ultrastructural changes and apoptosis of lung epithelial cells in broilers exposed to atmospheric ammonia. J Anim Sci 2021; 99:skab313. [PMID: 34718609 PMCID: PMC8599180 DOI: 10.1093/jas/skab313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/23/2021] [Indexed: 02/07/2023] Open
Abstract
To explore whether Radix Bupleuri extract (RBE) could protect lung injury of broilers under ammonia (NH3) exposure, 360 one-d-old male broilers were randomly allocated to four groups of six replicates each in a 2 × 2 factorial design with two diets (the basal diet [control; CON] and the basal diet supplemented with RBE [RB]) and two air conditions (normal condition [<2 ppm of NH3; NOR] and NH3 exposure [70 ppm of NH3; NH70]). The RB diet contained 80 mg saikosaponins/kg diet. On day 7, the lung tissues were collected and the lung epithelial cells (LEC) were isolated. Our experimental results showed that the NH3 exposure decreased body weight gain and feed intake irrespective of dietary treatments during days 1 to 7. However, the RBE addition decreased feed consumption to body weight gain ratio in broilers under NH70 conditions. In the LEC of CON-fed broilers under NH70 conditions, Golgi stacks showed the dilation of cisternaes and reduced secretory vesicles, mitochondria enlarged, the inner membrane of mitochondria became obscure, and the cristae of mitochondria ruptured, whereas only a mild enlargement of Golgi cisternaes and the part rupture of mitochondrial cristaes occurred in the LEC of RB-fed broilers under NH70 conditions. The NH3 exposure increased malondialdehyde (MDA) level, but decreased total antioxidant capacity (T-AOC) in the lungs of CON-fed broilers. However, the RBE addition decreased MDA level and increased T-AOC in the lungs of broilers under NH70 conditions. Simultaneously, the NH3 exposure increased apoptotic rate (AR), mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) level in the isolated LEC of CON-fed broilers. The RBE addition decreased AR, MMP, and ROS in the isolated LEC of broilers under NH70 condition. Besides, the NH3 exposure increased mRNA expression of B-cell lymphoma-2 associated X protein (BAX), caspase-3, and tumor necrosis factor α (TNF-α), but increased interferon γ (IFN-γ) mRNA abundance in the lungs of CON-fed broilers. The RBE supplement decreased mRNA levels of BAX, caspase-3, and TNF-α, but increased IFN-γ, interleukin (IL)-4, and IL-17 mRNA levels in the lungs of broilers under NH70 conditions. These results indicated that dietary RBE addition alleviated NH3 exposure-induced intercellular ultrastructural damage via mitochondrial apoptotic pathway, possibly due to RBE-induced increase of antioxidant capacity and immunomodulatory function in the lungs of broilers under NH3 exposure.
Collapse
Affiliation(s)
- Shiping Bai
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xi Peng
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Caimei Wu
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Tong Cai
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jiangfeng Liu
- School of Intelligence Technology, Geely University of China, Chengdu 641423, China
| | - Gang Shu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
38
|
Baicalin Inhibits NLRP3 Inflammasome Activity Via the AMPK Signaling Pathway to Alleviate Cerebral Ischemia-Reperfusion Injury. Inflammation 2021; 44:2091-2105. [PMID: 34080089 DOI: 10.1007/s10753-021-01486-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/30/2021] [Accepted: 05/23/2021] [Indexed: 02/07/2023]
Abstract
Baicalin has been reported to have ameliorative effects on nerve-induced hypoxic ischemia injury; however, its role in the NLRP3 inflammasome-dependent inflammatory response during cerebral ischemia-reperfusion remains unclear. To investigate the molecular mechanisms involved in baicalin alleviating cerebral ischemia-reperfusion injury, we investigated the AMPK signaling pathway which regulates NLRP3 inflammasome activity. SD rats were treated with baicalin at doses of 100 mg/kg and 200 mg/kg, respectively, after middle cerebral artery occlusion at 2 h and reperfusion for 24 h (MCAO/R). MCAO/R treatment significantly increased cerebral infarct volume, changed the ultrastructure of nerve cells, and activated the NLRP3 inflammasome, manifesting as significantly increased expression of NLRP3, ASC, cleaved caspase-1, IL-1β, and IL-18. Our results demonstrated that baicalin treatment effectively reversed these phenomena in a dose-dependent manner. Additionally, inhibition of NLRP3 expression was found to promote the neuroprotective effects of baicalin on cortical neurons. Furthermore, baicalin remarkably increased the expression of p-AMPK following oxygen glucose deprivation/reperfusion (OGD/R). The expression of the NLRP3 inflammasome was also increased when the AMPK pathway was blocked by compound C. Taken together, our findings reveal that baicalin reduces the activity of the NLRP3 inflammasome and consequently inhibits cerebral ischemia-reperfusion injury through activation of the AMPK signaling pathway.
Collapse
|
39
|
Li P, Hu S, Qian C, Yao Y, Li LY, Yang JF, Yang L, Yang CC, Zhou H, Wang SX, Hu Y, Zhu XY, Zhou J, Pan LX, Shen CP, Zhou H. The Therapeutic Effect of Traditional Chinese Medicine on Inflammatory Diseases Caused by Virus, Especially on Those Caused by COVID-19. Front Pharmacol 2021; 12:650425. [PMID: 34122073 PMCID: PMC8187915 DOI: 10.3389/fphar.2021.650425] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Inflammasomes are large multimolecular complexes best recognized because of their ability to control activation of caspase-1, which in turn regulates the maturation of interleukin-18 (IL-18) and interleukin-1 β (IL-1β). IL-1β was originally identified as a pro-inflammatory cytokine, capable of inducing local and systemic inflammation as well as a fever response reaction in response to infection or injury. Excessive production of IL-1β is related to inflammatory and autoimmune diseases. Both coronavirus disease 2019 (COVID-19) and severe acute respiratory syndrome (SARS) are characterized by excessive inflammatory response. For SARS, there is no correlation between viral load and worsening symptoms. However, there is no specific medicine which is available to treat the disease. As an important part of medical practice, TCM showed an obvious therapeutic effect in SARS-CoV-infected patients. In this article, we summarize the current applications of TCM in the treatment of COVID-19 patients. Herein, we also offer an insight into the underlying mechanisms of the therapeutic effects of TCM, as well as introduce new naturally occurring compounds with anti-coronavirus activity, in order to provide a new and potential drug development strategy for the treatment of COVID-19.
Collapse
Affiliation(s)
- Peng Li
- The First Affiliated Hospital of Medical University of Anhui, Hefei, China
| | - Shuang Hu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Cheng Qian
- Center for Scientific Research, Anhui Medical University, Hefei, China
| | - Yan Yao
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Liang-Yun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Jun-Fa Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Li Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Chen-Chen Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Hong Zhou
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Shu-Xian Wang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Ying Hu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Xing-Yu Zhu
- National Drug Clinical Trial Institution, The First Affiliated Hospital of Bengbu Medical College, Hefei, China
| | - Jing Zhou
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Lin-Xin Pan
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Chuan-Pu Shen
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Huan Zhou
- National Drug Clinical Trial Institution, The First Affiliated Hospital of Bengbu Medical College, Hefei, China
| |
Collapse
|
40
|
Yang F, Chen R, Li WY, Zhu HY, Chen XX, Hou ZF, Cao RS, Zang G, Li YX, Zhang W. D-Limonene Is a Potential Monoterpene to Inhibit PI3K/Akt/IKK-α/NF-κB p65 Signaling Pathway in Coronavirus Disease 2019 Pulmonary Fibrosis. Front Med (Lausanne) 2021; 8:591830. [PMID: 33768100 PMCID: PMC7985179 DOI: 10.3389/fmed.2021.591830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/27/2021] [Indexed: 12/31/2022] Open
Abstract
At the time of the prevalence of coronavirus disease 2019 (COVID-19), pulmonary fibrosis (PF) related to COVID-19 has become the main sequela. However, the mechanism of PF related to COVID (COVID-PF) is unknown. This study aimed to explore the key targets in the development of COVID-PF and the mechanism of d-limonene in the COVID-PF treatment. The differentially expressed genes of COVID-PF were downloaded from the GeneCards database, and their pathways were analyzed. d-Limonene was molecularly docked with related proteins to screen its pharmacological targets, and a rat lung fibrosis model was established to verify d-limonene's effect on COVID-PF-related targets. The results showed that the imbalance between collagen breakdown and metabolism, inflammatory response, and angiogenesis are the core processes of COVID-PF; and PI3K/AKT signaling pathways are the key targets of the treatment of COVID-PF. The ability of d-limonene to protect against PF induced by bleomycin in rats was reported. The mechanism is related to the binding of PI3K and NF-κB p65, and the inhibition of PI3K/Akt/IKK-α/NF-κB p65 signaling pathway expression and phosphorylation. These results confirmed the relationship between the PI3K–Akt signaling pathway and COVID-PF, showing that d-limonene has a potential therapeutic value for COVID-PF.
Collapse
Affiliation(s)
- Fan Yang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ru Chen
- Biomedical Research Institute of Fudan University, Shanghai, China
| | - Wan-Yang Li
- School of Public Health, Xiangya Medical College, Central South University, Changsha, China
| | - Hao-Yue Zhu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Xuan Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen-Feng Hou
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Ren-Shuang Cao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - GuoDong Zang
- Department of Pulmonary Diseases, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu-Xuan Li
- Second School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Zhang
- Department of Pulmonary Diseases, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
41
|
Fan J, Duan L, Wu N, Xu X, Xin J, Jiang S, Zhang C, Zhang H. Baicalin Ameliorates Pancreatic Fibrosis by Inhibiting the Activation of Pancreatic Stellate Cells in Mice with Chronic Pancreatitis. Front Pharmacol 2021; 11:607133. [PMID: 33536916 PMCID: PMC7848203 DOI: 10.3389/fphar.2020.607133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/02/2020] [Indexed: 02/05/2023] Open
Abstract
Pancreatic inflammation and fibrosis are typical pathological features in chronic pancreatitis (CP). Activated pancreatic stellate cells (PSCs) have been regarded as the core event in the development of pancreatic fibrosis and are considered to be the key target for treatment of CP. Baicalin (C21H18O11), the main chemical composition of Baikal skullcap in the traditional Chinese medicines Dachaihu decoction (DCHD) and Xiaochaihu decoction (XCHD), has shown significant effects in the treatment of pancreatic fibrosis in CP mice; however, whether baicalin can inhibit the activation of PSCs and its underlying mechanism remain unclear. In this study, the influence of baicalin on activated PSCs in vitro and in vivo was investigated, and the results showed that Baicalin could significantly ameliorate the degree of pancreatic inflammation and fibrosis, while decreasing the levels of alpha-smooth muscle actin (α-SMA), F4/80 (surface markers of mouse macrophages), nuclear factor kappa-B (NF-κB), monocyte chemotactic protein 1 (MCP-1), and collagen type I alpha 1 (COL1A1)in the pancreas. Moreover, NF-κB and α-SMA were co-expressed in the pancreas of CP mice. Baicalin treatment markedly reduced the expression of co-location of α-SMA and NF-κB. In vitro, the protein expression levels of transforming growth factor-β receptor 1 (TGF-βR1), phosphorylated TGF-β activated kinase 1 p-TAK 1, and NF-κBp65 in PSCs were all remarkably reduced after treatment with baicalin. In addition, baicalin could inhibit MCP-1 mRNA expression in supernatant of activated PSCs, as well as the excessive migration of macrophages. Taken together, our findings indicated that baicalin could inhibit the TGF-β1/TGF-βR1/TAK1/NF-κB signaling pathway of activated PSCs, reduce the secretion of MCP-1, and further decrease the infiltration of macrophages and inflammation cells of the local microenvironment of the pancreas. Thus, this study provides a reliable experimental basis for baicalin in the prevention and treatment of CP.
Collapse
Affiliation(s)
- Jianwei Fan
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lifang Duan
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Nan Wu
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaofan Xu
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jiaqi Xin
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Shengnan Jiang
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Cheng Zhang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital, Xianyang, China
| | - Hong Zhang
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|