1
|
Tang R, Zhang L, Lou J, Mo W, Zhao L, Li L, Zhang K, Yu Q. Formononetin prevents intestinal injury caused by radiotherapy in colorectal cancer mice via the Keap1-Nrf2 signaling pathway. Biochem Biophys Res Commun 2025; 761:151676. [PMID: 40184790 DOI: 10.1016/j.bbrc.2025.151676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/20/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025]
Abstract
Radiation-induced intestinal injury (RIII) is a serious complication of radiation therapy. Formononetin (FT) is a methoxy isoflavone with anti-tumor and anti-oxidant properties. This study explored whether FT reduces ROS by activating the Keap1/Nrf2 pathway and thereby protecting against RIII. 30 BABL/C mice were used to construct a colorectal cancer model with CT26 cells and validated the model on day 7. Then, the cancer model mice were divided into 3 groups on day 14 (model control, model, and model + FT) with administration of FT or saline and radiated them on day 21. All samples were collected on day 28. Mechanical intestinal barrier assessed via intestinal absorption function, HE staining, and Claudin-1, Occludin, ZO-1 expression. Apoptosis was measured by TUNEL and cleaved-caspase 3, angiogenesis by CD31, and oxidative stress by NO and ROS levels. Keap1, Nrf2 (cytoplasm) and Nrf2 (nucleus) expression were measured by Western blot. FT treatment enhanced absorption and reduced damage and apoptosis in intestinal tissue. During FT treatment, inhibition of NO and ROS leads to suppression of oxidative stress and promotion of CD31 expression promotes angiogenesis. FT upregulated the expression of nuclear Nrf2 and reduced cytoplasmic Keap1 and Nrf2. FT strengthens mechanical barrier and decreases oxidative stress in intestinal tissue, providing radiation injury protection. Its anti-radiation effect may be through Keap1/Nrf2 pathway activation.
Collapse
Affiliation(s)
- Rongjun Tang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lidan Zhang
- Radiotherapy Center, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jun Lou
- Department of Ultrasound, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Weixing Mo
- Department of Radiology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Linfang Zhao
- Department of Ultrasound, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lingdi Li
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ke Zhang
- Department of Radiotherapy, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Qingqing Yu
- Department of Radiotherapy, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Gould S, Herron A, Davis J, Phillips M, Chakrabarti M, Evans CE. Control of inflammatory lung injury and repair by metabolic signaling in endothelial cells. Curr Opin Hematol 2025; 32:157-167. [PMID: 39450949 PMCID: PMC11949724 DOI: 10.1097/moh.0000000000000848] [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] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW Sepsis-induced inflammatory lung injury includes acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). There are currently no effective treatments for ALI/ARDS, but clinical outcomes could be improved by inhibiting lung injury and/or promoting post-sepsis vascular repair. In this review, we describe studies of endothelial cell metabolic pathways in sepsis-induced ALI/ARDS and vascular repair and identify areas of research that deserve attention in future studies. We also describe studies of metabolic interventions that aim to inhibit ALI/ARDS and/or promote post-sepsis vascular repair, including those that target endothelial cell metabolites, endothelial cell metabolic signaling pathways, and endothelial cell metabolism. RECENT FINDINGS Endothelial cells are integral to both the injury and repair phases of ALI/ARDS. During the injury phase of ALI/ARDS, lung endothelial cell survival decreases, and lung endothelial cell-to-endothelial cell (EC-EC) junctions are weakened. During the repair phase after sepsis-induced lung injury, lung endothelial cell proliferation and lung EC-EC junction reannealing occur. These crucial aspects of ALI/ARDS and post-sepsis vascular repair, that is, endothelial cell viability, growth, and junction integrity, are controlled by a myriad of metabolites and metabolic signaling pathways in endothelial cells. SUMMARY Metabolic signaling pathways in endothelial cells represent a novel class of putative targets for the prevention and treatment of sepsis-induced inflammatory lung injury. Therapies that target metabolic signaling in endothelial cells are currently being explored as potential treatments for sepsis-induced inflammatory lung injury.
Collapse
Affiliation(s)
- Seth Gould
- Biomedical Engineering Program, University of South Carolina
| | - Ansley Herron
- Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina
| | - Jonathan Davis
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Mollie Phillips
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Mrinmay Chakrabarti
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
| | - Colin E. Evans
- Biomedical Engineering Program, University of South Carolina
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine
- Institute on Cardiovascular Disease Research, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
3
|
Xing A, Wang F, Liu J, Zhang Y, He J, Zhao B, Sun B. The prospect and underlying mechanisms of Chinese medicine in treating periodontitis. Chin J Nat Med 2025; 23:269-285. [PMID: 40122658 DOI: 10.1016/s1875-5364(25)60842-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/18/2024] [Accepted: 07/13/2024] [Indexed: 03/25/2025]
Abstract
Inflammation represents a critical immune response triggered by cellular activities and inflammatory mediators following tissue damage. It plays a central role in the pathological progression of diverse diseases, including psychiatric disorders, cancer, and immunological conditions, rendering it an essential target for therapeutic intervention. Periodontitis, a prevalent oral inflammatory disease, is a leading cause of tooth loss and poses significant health challenges globally. Traditionally, inflammatory diseases such as periodontitis have been treated with systemic administration of synthetic chemicals. However, recent years have witnessed challenges, including drug resistance and microbial dysbiosis associated with these treatments. In contrast, natural products derived from Chinese medicine offer numerous benefits, such as high safety profiles, minimal side effects, innovative pharmacological mechanisms, ease of extraction, and multiple targets, rendering them viable alternatives to conventional antibiotics for treating inflammatory conditions. Numerous effective anti-inflammatory natural products have been identified in traditional Chinese medicine (TCM), including alkaloids, flavonoids, terpenoids, lignans, and other natural products that exhibit inhibitory effects on inflammation and are potential therapeutic agents. Several studies have confirmed the substantial anti-inflammatory and immunomodulatory properties of these compounds. This comprehensive review examines the literature on the anti-inflammatory effects of TCM-derived natural products from databases such as PubMed, Web of Science, and CNKI, focusing on terms like "inflammation", "periodontitis", "pharmacology", and "traditional Chinese medicine". The analysis systematically summarizes the molecular pharmacology, chemical composition, and biological activities of these compounds in inflammatory responses, alongside their mechanisms of action. This research seeks to deepen understanding of the mechanisms and biological activities of herbal extracts in managing inflammatory diseases, potentially leading to the development of promising new anti-inflammatory drug candidates. Future applications could extend to the treatment of various inflammatory conditions, including periodontitis.
Collapse
Affiliation(s)
- Aili Xing
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Feng Wang
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Jinzhong Liu
- Preventive Dentistry, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Yuan Zhang
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Jingya He
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China
| | - Bin Zhao
- Periodontics, Hospital of Stomatologyl, Jilin University, Changchun 130021, China.
| | - Bin Sun
- Oral and Maxillofacial Surgery, Hospital of Stomatologyl, Jilin University, Changchun 130021, China.
| |
Collapse
|
4
|
Asci H, Emre Akin S, Ekrem Camas H, Bindal A, Kurtbolat O, Tasan S, Gulal A, Taner R, Kurt T, Ozmen O. Investigating the protective effects of fluvoxamine against sepsis-related acute lung injury through antiapoptotic, anti-inflammatory, and anti-oxidant features in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:323-331. [PMID: 39906615 PMCID: PMC11790192 DOI: 10.22038/ijbms.2024.80608.17444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/12/2024] [Indexed: 02/06/2025]
Abstract
Objectives Acute lung injury (ALI) is characterized by severe hypoxia and alveolar damage, often caused by oxidative stress, endoplasmic reticulum stress (ERS), and apoptosis. Fluvoxamine (FLV), an antidepressant, has tissue-protective properties through various intracellular mechanisms. This study investigates the anti-inflammatory effects of FLV used as an antidepressant in a lipopolysaccharide (LPS)-induced ALI model. Materials and Methods Thirty-two female Wistar Albino rats aged 14-16 weeks and weighing 300-350 g, with 8 animals in each group, were divided into four groups: control, LPS, LPS+FLV, and FLV. After LPS administration, rats were euthanized, and histopathological analysis, immunohistochemistry for tumor necrosis factor-α (TNF-α) and caspase-3 (Cas-3), ELISA for oxidative stress markers, and PCR for CHOP, Cas-12, and Cas-9 gene expressions were conducted. Results In the LPS group, lung tissue damage, increased inflammatory cell infiltration, increased Cas-3 and TNF-α expressions, increased oxidative stress markers, and increased CHOP, Cas-9, and Cas-12 mRNA expressions were observed compared to the control group. FLV treatment in the LPS+FLV group significantly reversed these effects in the LPS group. Conclusion FLV exhibits protective effects against ALI by mitigating inflammation, ERS, and apoptosis via the CHOP/Cas-9/Cas-12 pathway. Further studies are needed to explore additional pathways and potential clinical applications of FLV.
Collapse
Affiliation(s)
- Halil Asci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
- Department of Bioengineering, Institute of Science and Technology, Suleyman Demirel University Isparta, Turkey
| | - Suleyman Emre Akin
- Department of Thoracic Surgery, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Hasan Ekrem Camas
- Department of Thoracic Surgery, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Ahmet Bindal
- Department of Anesthesiology and Reanimation, Suleyman Demirel University, Isparta, Turkey
| | - Okan Kurtbolat
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Serife Tasan
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | | | - Rumeysa Taner
- Department of Bioengineering, Institute of Science and Technology, Suleyman Demirel University Isparta, Turkey
| | - Turgut Kurt
- Veterinary Technician, Faculty of Medicine, Suleyman Demirel University Isparta, Turkey
| | - Ozlem Ozmen
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
5
|
Wu Y, Feng K, Chen Y, Zhang H, Zhang M, Han B, Chen X, Yang L, Wang X, Li W, Tang J. Exploring the anti-inflammatory and immunomodulatory potential of licochalcone B against psoralidin-induced liver injury. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118600. [PMID: 39053714 DOI: 10.1016/j.jep.2024.118600] [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: 01/29/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herb-induced liver injury (HILI) represents an exacerbated inflammatory response, with Psoraleae fructus (PF) and its preparations recently associated with hepatotoxicity. Licorice, historically recognized as a detoxifying herbal remedy, is considered to possess hepatoprotective properties. Our previous research identified bavachin, bakuchiol, and psoralidin (PSO) as potential toxic constituents in PF, while licochalcone B (LCB) and echinatin were identified as bioactive components in licorice. However, evidence regarding the interactions of active compounds in herbs and their underlying mechanisms remains limited. AIM OF THE STUDY The objective of this study is to assess the potential mechanisms through which LCB modulates immunological and anti-inflammatory responses to treat PSO-induced liver injury by using human hepatocyte cells (L02) and LPS-primed mice. METHODS The ameliorative effects of LCB and echinatin on bavachin, bakuchiol, and PSO-induced liver injury were demonstrated in L02 cells. Subsequently, the efficacy of LCB on PSO-induced idiosyncratic liver injury was further validated in C57BL/6 mice under moderate inflammatory stress induced by LPS priming. The mechanisms were preliminarily explored with an integrated strategy of molecular docking, RT-PCR verification, and untargeted metabolomics. RESULTS The study shows that LCB significantly reduced cell injury induced by the three chemicals in PF and provided substantial protection against PSO-induced hepatic damage, as indicated by the levels of ALT, AST, and LDH. LCB normalized liver function and remarkedly alleviated hepatic lesions and inflammation caused by PSO in mice under moderate inflammatory stress. The mRNA profiles of both L02 cells and mice liver tissue revealed that LCB mitigated PSO-induced hepatotoxicity by regulating the gene expression of pro-inflammatory cytokines IL1B and TNF, as well as immunoinflammatory genes PIK3CA, AKT1, NFKB1, and NLRP3. Furthermore, untargeted metabolomics of liver tissue indicated that LCB could reverse the abnormal expression of 11 discriminatory metabolites, with the interrelationship between differential metabolites and target genes primarily clustering in glycerophospholipid metabolism, arachidonic acid metabolism, and phosphatidylinositol signaling system. CONCLUSION LCB demonstrated a superior anti-inflammatory and immunomodulatory effect on PSO-induced hepatotoxicity by modulating the inflammatory response and metabolic signaling system. Key interactive targets included phosphatidylcholine, phosphatidic acid, and subunit isoforms of PI3K.
Collapse
Affiliation(s)
- Yali Wu
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China
| | - Keran Feng
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China
| | - Yulong Chen
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; College Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Zhang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China
| | - Mingliang Zhang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China
| | - Bing Han
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaofei Chen
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China
| | - Liuqing Yang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China
| | - Xiaoyan Wang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China.
| | - Weixia Li
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China.
| | - Jinfa Tang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450003, China; Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, 450003, China; School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China.
| |
Collapse
|
6
|
Wang X, Gao M, Lu X, Lei Y, Sun J, Ren M, Xu T, Lin H. Resveratrol alleviates Mono-2-ethylhexyl phthalate-induced mitophagy, ferroptosis, and immunological dysfunction in grass carp hepatocytes by regulating the Nrf2 pathway. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 371:123235. [PMID: 39509968 DOI: 10.1016/j.jenvman.2024.123235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Mono-2-ethylhexyl phthalate (MEHP) is the major biologically active metabolite of Di(2-ethylhexyl) phthalate (DEHP). This MEHP mono-ester metabolite can be transported through the bloodstream into tissues such as the liver, kidneys, fat, and testes and cause corresponding damage. Resveratrol (RSV) has anti-inflammatory, antioxidant, and detoxification characteristics. Our research examined whether RSV alleviates MEHP-induced grass carp hepatocyte (L8824 cell) injury and its relationship with the Nrf2 pathway, mitophagy, ferroptosis, and immune function. Therefore, we treated L8824 cells with 85 μM MEHP and/or 2 μM RSV. The findings indicated that exposing MEHP resulted in increased reactive oxygen species (ROS) content and decreased mitochondrial membrane potential in L8824 cells, which induced an up-regulation of the expression of mitophagy-related indicators (PINK1, Parkin, Beclin1, LC3B, and ATG5) and a down-regulation of P62. An up-regulation of the expression of the ferroptosis-related indicators TFR1 and COX-2, and GPX4 and FTH expression was down-regulated. In addition, there was a decrease in the expression of IL-2 and IFN-γ and an increase in the expression of inflammatory cytokines such as TNF-α, IL-1β, and IL-6 after exposure to MEHP. RSV activates the Nrf2 pathway and effectively alleviates MEHP-induced mitophagy, ferroptosis, and immunologic dysfunction of L8824 cells.
Collapse
Affiliation(s)
- Xiaodan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Meichen Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiunan Lu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yutian Lei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jiatong Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Mengyao Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tong Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Hongjin Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
7
|
Wang Q, Jin Q, Wang F, Wang Y, Li X, Zhou Y, Xu S, Fu A, Li W. Bacillus amyloliquefaciens SC06 alleviates LPS-induced intestinal damage by inhibiting endoplasmic reticulum stress and mitochondrial dysfunction in piglets. Int J Biol Macromol 2024; 282:137307. [PMID: 39510464 DOI: 10.1016/j.ijbiomac.2024.137307] [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: 11/12/2023] [Revised: 09/17/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Endoplasmic reticulum stress (ERS) and mitochondrial dysfunction play an important role in the pathogenesis of intestinal diseases. Our studies investigated the effects of Bacillus amyloliquefaciens SC06 on jejunal mitochondria and ER in piglets under the LPS-induced intestinal injury model. Eighteen piglets (male, 21 days old) were randomly assigned to three treatments: CON (basal diet), LPS (basal diet +100 μg/kg LPS), and SC06 + LPS (basal diet +1 × 108 cfu/kg SC06 + 100 μg/kg LPS). Compared to the LPS group, administration of SC06 improved jejunal morphology and barrier function. In addition, SC06 reduced reactive oxygen species (ROS) and MDA generation in the jejunum by activating the Nrf2/keap1 pathway, which increased the activity of CAT, GSH and SOD in LPS-challenged pigs. In addition, SC06 reduced LPS-induced mitochondrial dysfunction and ERS as evidenced by a decrease in ROS, an improvement in mitochondrial membrane potential and an increase in adenosine triphosphate levels. The results of in vitro IPEC-J2 cell experiments also indicate that SC06 can reduce LPS-induced oxidative stress, mitochondrial dysfunction, ERS, and intestinal barrier function damage by activating the Nrf2/keap1 signaling pathway. Finally, treatment with the Nrf2-specific inhibitor ML-385 inhibited the upregulated effect of SC06 on antioxidant capacity and intestinal barrier function in IPEC-J2 cells. In conclusion, SC06 ameliorated intestinal damage and mitochondrial dysfunction and attenuated endoplasmic reticulum stress via activation of the Nrf2/keap1 signaling pathway in LPS-challenged piglets.
Collapse
Affiliation(s)
- Qi Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qian Jin
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fei Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yang Wang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiang Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanhao Zhou
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shujie Xu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Aikun Fu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Weifen Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
8
|
Zhang M, Wang J, Liu R, Wang Q, Qin S, Chen Y, Li W. The role of Keap1-Nrf2 signaling pathway in the treatment of respiratory diseases and the research progress on targeted drugs. Heliyon 2024; 10:e37326. [PMID: 39309822 PMCID: PMC11414506 DOI: 10.1016/j.heliyon.2024.e37326] [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: 05/28/2024] [Revised: 07/30/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Lungs are exposed to external oxidants from the environment as in harmful particles and smog, causing oxidative stress in the lungs and consequently respiratory ailment. The NF-E2-related factor 2 (Nrf2) is the one with transcriptional regulatory function, while its related protein Kelch-like ECH-associated protein 1 (Keap1) inhibits Nrf2 activity. Together, they form the Keap1-Nrf2 pathway, which regulates the body's defense against oxidative stress. This pathway has been shown to maintain cellular homeostasis during oxidative stressing, inflammation, oncogenesis, and apoptosis by coordinating the expression of cytoprotective genes and making it a potential therapeutic target for respiratory diseases. This paper summarizes this point in detail in Chapter 2. In addition, this article summarizes the current drug development and clinical research progress related to the Keap1-Nrf2 signaling pathway, with a focus on the potential of Nrf2 agonists in treating respiratory diseases. Overall, the article reviews the regulatory mechanisms of the Keap1-Nrf2 signaling pathway in respiratory diseases and the progress of targeted drug research, aiming to provide new insights for treatment.
Collapse
Affiliation(s)
- Mengyang Zhang
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Jing Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Runze Liu
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Qi Wang
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Song Qin
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, 92093, USA
| | - Wenjun Li
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| |
Collapse
|
9
|
Samutrtai P, Yingchutrakul Y, Faikhruea K, Vilaivan T, Chaikeeratisak V, Chatwichien J, Krobthong S, Aonbangkhen C. Vernonia amygdalina Leaf Extract Induces Apoptosis in HeLa Cells: A Metabolomics and Proteomics Study. Pharmaceuticals (Basel) 2024; 17:1079. [PMID: 39204184 PMCID: PMC11360076 DOI: 10.3390/ph17081079] [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: 07/29/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Medicinal plants produce various bioactive molecules with potential anti-cancer properties with favorable safety profiles. We aimed to investigate the comprehensive composition of Vernonia amygdalina leaf extract and its cytotoxic effects via apoptosis in HeLa cells. The metabolomics approach using LC-MS/MS was conducted to gather the metabolite profile of the extract. Proteomics was performed to understand the comprehensive mechanistic pathways of action. The apoptosis was visualized by cellular staining and the apoptotic proteins were evaluated. V. amygdalina leaf extract exhibited dose-dependent cytotoxic effects on both HeLa and Vero cells after 24 h of exposure in the MTT assay with the IC50 values of 0.767 ± 0.0334 and 4.043 ± 0.469 µg mL-1, respectively, which demonstrated a higher concentration required for Vero cell cytotoxicity. The metabolomic profile of 112 known metabolites specified that the majority of them were alkaloids, phenolic compounds, and steroids. Among these metabolites, deacetylvindoline and licochalcone B were suggested to implicate cytotoxicity. The cytotoxic pathways involved the response to stress and cell death which was similar to doxorubicin. The upstream regulatory proteins, phosphatase and tensin homolog deleted on chromosome ten (PTEN) and X-box binding protein 1 (XBP1), were significantly altered, supporting the regulation of apoptosis and cell death. The levels of apoptotic proteins, c-Jun N-terminal kinases (JNK), p53, and caspase-9 were significantly increased. The novel insights gained from the metabolomic profiling and proteomic pathway analysis of V. amygdalina leaf extract have identified crucial components related to apoptosis induction, highlighting its potential to develop future chemotherapy.
Collapse
Affiliation(s)
- Pawitrabhorn Samutrtai
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Yodying Yingchutrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand;
| | - Kriangsak Faikhruea
- Organic Synthesis Research Unit (OSRU), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (K.F.); (T.V.)
| | - Tirayut Vilaivan
- Organic Synthesis Research Unit (OSRU), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (K.F.); (T.V.)
| | - Vorrapon Chaikeeratisak
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Jaruwan Chatwichien
- Program in Chemical Sciences, Chulabhorn Graduate Institute (CGI), Bangkok 10210, Thailand;
| | - Sucheewin Krobthong
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (C.A.)
| | - Chanat Aonbangkhen
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (C.A.)
- Center of Excellence on Petrochemical and Materials Technology, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
10
|
Guo B, Wang H, Zhang Y, Wang C, Zhang H, Zhao Y, Qin J. Glycyrrhizin alleviates BoAHV-1-induced lung injury in guinea pigs by inhibiting the NF-κB/NLRP3 Signaling pathway and activating the Nrf2/HO-1 Signaling pathway. Vet Res Commun 2024; 48:2499-2511. [PMID: 38865040 DOI: 10.1007/s11259-024-10436-7] [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: 11/28/2023] [Accepted: 06/10/2024] [Indexed: 06/13/2024]
Abstract
Varicellovirus bovinealpha 1 (BoAHV-1) is a significant pathogen responsible for respiratory disease in cattle, capable of inducing lung damage independently or co-infection with bacteria. The widespread spread of BoAHV-1 in cattle herds has caused substantial economic losses to the cattle industry. The pathogenic mechanisms of BoAHV-1 are often relevant to robust inflammatory responses, increased oxidative burden, and the initiation of apoptosis. Glycyrrhizin (GLY) is a small-molecule triterpenoid saponin compound obtained from the herb liquorice, which has a broad spectrum of pharmacological properties such as antiviral, anti-inflammatory, and antioxidant effects. Furthermore, GLY regulates lung physiology by modulating oxidative stress, inflammatory response, and cell apoptosis through interference with the NF-κB/NLRP3 and Nrf2/HO-1 Signaling pathways. However, the potential of GLY to mitigate lung injury induced by BoAHV-1 and its underlying mechanism remains unclear. Therefore, in this study, we investigated the protective effect of GLY against pulmonary injury induced by BoAHV-1 in a guinea pig model by reducing viral load and suppressing the inflammatory response, oxidative stress, and apoptosis. The results of this study demonstrated that GLY exerted a protective effect against BoAHV-1-induced lung injury in guinea pigs. Specifically, GLY reduced the levels of pro-inflammatory cytokines interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and interleukin (IL)-8 in guinea pig tissues while suppressing the expression of Caspase-1. Additionally, GLY reduced BoAHV-1 load and the number of TUNEL-positive lung cells in guinea pig lungs while inhibiting Caspase 3 protein expression. Furthermore, GLY significantly enhanced lung antioxidant capacity by increasing superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) activity while simultaneously reducing malondialdehyde (MDA) levels. Lung histological observation and score further validated the protective effect of GLY on BoAHV-1-induced lung injury. Furthermore, we observed that the expression of phosphorylated NF-κB p65 (p-NF-κB p65) and NLRP3 proteins in the lung tissue of BoAHV-1-infected guinea pigs decreased after GLY treatment while the expression of Nrf2 and HO-1 proteins increased. These results indicated that GLY inhibited the NF-κB/NLRP3 Signaling pathway and activated the Nrf2/HO-1 Signaling pathway during BoAHV-1 infection. Ultimately, our findings demonstrated that GLY alleviates BoAHV-1-induced inflammation response, oxidative stress, and cell apoptosis by inhibiting the NF-κB/NLRP3 Signaling pathway and activating the Nrf2/HO-1 Signaling pathway to protect guinea pigs from lung injury caused by BoAHV-1. Ultimately, our findings demonstrated that GLY alleviates BoAHV-1-induced inflammation response, oxidative stress, and cell apoptosis by inhibiting the NF-κB/NLRP3 Signaling pathway and activating the Nrf2/HO-1 Signaling pathway to protect guinea pigs from lung injury caused by BoAHV-1. Importantly, this study provides a compelling argument for the GLY in combating respiratory disease in cattle caused by BoAHV-1.
Collapse
Affiliation(s)
- Bing Guo
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei Province, China
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Haifeng Wang
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Yue Zhang
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei Province, China
| | - Chuanwen Wang
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei Province, China
| | - Huaying Zhang
- Hebei Provincial Animal Science and Veterinary Institute, Shijiazhuang, Hebei Province, China
| | - Yian Zhao
- Zhangjiakou Rural Cooperative Economy Business Management Station, Zhangjiakou, Hebei Province, China
| | - Jianhua Qin
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei Province, China.
| |
Collapse
|
11
|
Lei X, Liu X, Yu J, Li K, Xia L, Su S, Lin P, Zhang D, Li Y. 3-methyladenine ameliorates acute lung injury by inhibiting oxidative damage and apoptosis. Heliyon 2024; 10:e33996. [PMID: 39055838 PMCID: PMC11269838 DOI: 10.1016/j.heliyon.2024.e33996] [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: 01/04/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Background Acute lung injury (ALI) is a condition characterized by inflammation and oxidative damage. 3-methyladenine (3-MA) has great potential for regulating apoptosis, but its regulatory role in ALI is unknown. Methods Lipopolysaccharide (LPS)-treated mice and tert-butyl hydroperoxide (TBHP)-treated bronchial epithelial cells were used to simulate in vivo and in vitro ALI models, respectively. In vivo, lung injury was assessed by histopathological analysis and lung injury scoring. The total cell count, protein content, and inflammatory factors in bronchoalveolar lavage fluid (BALF) were examined. The level of apoptosis in lung tissue was assessed through TUNEL staining. In the vitro ALI model, cell viability and levels of reactive oxygen species and apoptosis were assessed. Results 3-MA pretreatment ameliorated lung injury, including intra-alveolar hemorrhage and inflammatory cell accumulation, both in vitro and in vivo. 3-MA pretreatment also decreased inflammatory factor levels in the BALF. 3-MA pretreatment alleviated oxidative damage, decreased reactive oxygen species levels, and attenuated morphological changes. TUNEL and Annexin V-FITC/PI staining revealed that pretreatment with 3-MA reduced the level of apoptosis. 3-MA pretreatment significantly decreased the expression of caspase-3 and Bax but increased the expression of Bcl-2 in ALI. Mechanistically, 3-MA pretreatment also affected the PKCα/NOX4 and Nrf2 pathways, which decreased the level of apoptosis in ALI. Conclusions 3-MA pretreatment inhibited inflammation and oxidative damage in ALI and inhibited apoptosis to mitigate ALI in part by inhibiting the PKCα/NOX4 pathway and activating the Nrf2 pathway. Based on these results, 3-MA might be a viable medication to treat with ALI.
Collapse
Affiliation(s)
- Xiong Lei
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiling Liu
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia Yu
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kai Li
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Lijing Xia
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shanshan Su
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pengcheng Lin
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dan Zhang
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuping Li
- The Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Shaikh S, Lee EJ, Ahmad K, Choi I. Therapeutic potential and action mechanisms of licochalcone B: a mini review. Front Mol Biosci 2024; 11:1440132. [PMID: 39021879 PMCID: PMC11251949 DOI: 10.3389/fmolb.2024.1440132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Licochalcone B (LicB), a chalcone derived from Glycyrrhiza uralensis and Glycyrrhiza glabra, has received considerable attention due to its diverse pharmacological properties. Accumulated data indicates that LicB has pharmacological effects that include anti-cancer, hepatoprotective, anti-inflammatory, and neuroprotective properties. The action mechanism of LicB has been linked to several molecular targets, such as phosphoinositide 3-kinase/Akt/mammalian target of rapamycin, p53, nuclear factor-κB, and p38, and the involvements of caspases, apoptosis, mitogen-activated protein kinase-associated inflammatory pathways, and anti-inflammatory nuclear factor erythroid 2-related factor 2 signaling pathways highlight the multifaceted therapeutic potential of LicB. This review systematically updates recent findings regarding the pharmacological effects of LicB, and the mechanistic pathways involved, and highlights the potential use of LicB as a promising lead compound for drug discovery.
Collapse
Affiliation(s)
- Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
13
|
Luo C, Ye Y, Lv A, Zuo W, Yang Y, Jiang C, Ke J. The impact of Astragaloside IV on the inflammatory response and gut microbiota in cases of acute lung injury is examined through the utilization of the PI3K/AKT/mTOR pathway. PLoS One 2024; 19:e0305058. [PMID: 38954702 PMCID: PMC11218977 DOI: 10.1371/journal.pone.0305058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/22/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVES Astragaloside IV (AS-IV) is a natural triterpenoid saponin compound with a variety of pharmacological effects, and several studies have clarified its anti-inflammatory effects, which may make it an effective alternative treatment against inflammation. In the study, we aimed to investigate whether AS-IV could attenuate the inflammatory response to acute lung injury and its mechanisms. METHODS Different doses of AS-IV (20mg·kg-1, 40mg·kg-1, and 80mg·kg-1) were administered to the ALI rat model, followed by collection of serum and broncho alveolar lavage fluid (BALF) for examination of the inflammatory response, and HE staining of the lung and colon tissues, and interpretation of the potential molecular mechanisms by quantitative real-time PCR (qRT-PCR), Western blotting (WB). In addition, fecal samples from ALI rats were collected and analyzed by 16S rRNA sequencing. RESULTS AS-IV decreased the levels of TNF-α, IL-6, and IL-1β in serum and BALF of mice with Acute lung injury (ALI). Lung and colon histopathology confirmed that AS-IV alleviated inflammatory infiltration, tissue edema, and structural changes. qRT-PCR and WB showed that AS-IV mainly improved inflammation by inhibiting the expression of PI3K, AKT and mTOR mRNA, and improved the disorder of intestinal microflora by increasing the number of beneficial bacteria and reducing the number of harmful bacteria. CONCLUSION AS-IV reduces the expression of inflammatory factors by inhibiting the PI3K/AKT/mTOR pathway and optimizes the composition of the gut microflora in AIL rats.
Collapse
Affiliation(s)
- Cheng Luo
- Clinical College of traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yuanhang Ye
- Clinical College of traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Anqi Lv
- Clinical College of traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Wanzhao Zuo
- Clinical College of traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yi Yang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, China
| | - Cheng Jiang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, China
| | - Jia Ke
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei Academy of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
14
|
Cai L, Xu L, Shen K, Wang Q, Ni R, Xu X, Ma X. Sophorae tonkinensis radix polysaccharide attenuates acetaminophen-induced liver injury by regulating the miR-140-5p-related antioxidant mechanism. J Tradit Complement Med 2024; 14:467-476. [PMID: 39035693 PMCID: PMC11259709 DOI: 10.1016/j.jtcme.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/14/2023] [Accepted: 01/16/2024] [Indexed: 07/23/2024] Open
Abstract
STRP1, a polysaccharide active ingredient isolated from the traditional Chinese medicine Sophorae tonkinensis radix, has demonstrated a protective effect against acetaminophen (APAP)-induced liver injury (AILI). The underlying molecular mechanism was investigated in this study. Here, an acute liver damage mouse model was generated by APAP (400 mg/kg) and used to identify the protective effect of STRP1 (200 mg/kg) on mouse livers. In vitro cell experiments were used to further verify the related signaling pathways. Initially, in our study, STRP1 treatment reduced APAP-induced liver injury by decreasing aminotransferase activity and cell apoptosis and increasing cell proliferation. Furthermore, STRP1 treatment significantly increased Nrf2 expression and alleviated oxidative stress caused by reactive oxygen species in AILI. Based on bioinformatics and experimental studies, miR-140-5p was identified and found to be reduced by STRP1, increasing Nrf2 expression. Additionally, Nrf2 played an important role in the protective impact of STRP1-suppressed miR-140-5p expression. Generally, these results showed that STRP1-mediated suppression of miR-140-5p expression mitigates AILI by activating the Nrf2-mediated Nrf2-Keap1 pathway. This study revealed that STRP1 might be a potential treatment agent for AILI.
Collapse
Affiliation(s)
- Liangliang Cai
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, PR China
| | - Lixing Xu
- School of Pharmacy, Jiangsu Key Laboratory of Inflammation and Molecular Drug Targets, Nantong University, Nantong, 226001, PR China
| | - Kai Shen
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, PR China
| | - Qin Wang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, PR China
| | - Ronghua Ni
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, PR China
| | - Xin Xu
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, PR China
| | - Xiaofei Ma
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, PR China
| |
Collapse
|
15
|
Wei CY, Zhang X, Si LN, Shu WH, Jiang SN, Ding PJ, Cheng LY, Sun TC, Yang SH. Melatonin activates Nrf2/HO-1 signalling pathway to antagonizes oxidative stress-induced injury via melatonin receptor 1 (MT1) in cryopreserved mice ovarian tissue. Reprod Domest Anim 2024; 59:e14598. [PMID: 38881434 DOI: 10.1111/rda.14598] [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: 02/22/2024] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 06/18/2024]
Abstract
Our previous research has shown that melatonin (MLT) can reduce cryopreserved ovarian damage in mice. Yet, the molecular mechanism of MLT protection is still unclear. Some studies have shown that melatonin receptor 1 (MT1) is very important for animal reproductive system. To evaluate whether MLT exerts its protective effect on cryopreserved mice ovarian tissue via MT1, we added antagonist of MT1/MT2 (Luzindor) or antagonist of MT2 (4P-PDOT) to the freezing solution, followed by cryopreservation and thawing of ovarian tissue. The levels of total superoxide dismutase (T-SOD), catalase (CAT), nitric oxide (NO) and malondialdehyde (MDA) were detected. Besides, by using RT-PCR and Western blotting, the expression of Bcl-2, Bax and Nrf2/HO-1 signalling pathway-related proteins was detected. These findings demonstrated that compared with the melatonin group, the addition of Luzindor increased apoptosis, NO and MDA activities, decreased CAT and T-SOD activities and inhibited Nrf2/HO-1 signalling pathway. In conclusion, melatonin can play a protective role in cryopreserved ovarian tissue of mice through MT1 receptor.
Collapse
Affiliation(s)
- Chen Yang Wei
- Faculty of Graduate Studies, Chengde Medical University, Chengde, Hebei, China
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei, China
| | - Xin Zhang
- Faculty of Graduate Studies, Chengde Medical University, Chengde, Hebei, China
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei, China
| | - Li Na Si
- Faculty of Graduate Studies, Chengde Medical University, Chengde, Hebei, China
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei, China
| | - Wei Han Shu
- Faculty of Graduate Studies, Chengde Medical University, Chengde, Hebei, China
- Department of Immunology, Chengde Medical University, Chengde, Hebei, China
| | - Sheng Nan Jiang
- Faculty of Graduate Studies, Chengde Medical University, Chengde, Hebei, China
- Department of Immunology, Chengde Medical University, Chengde, Hebei, China
| | - Pei Jian Ding
- Department of Gastrointestinal Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| | - Lu Yang Cheng
- Faculty of Graduate Studies, Chengde Medical University, Chengde, Hebei, China
- Department of Immunology, Chengde Medical University, Chengde, Hebei, China
| | - Tie Cheng Sun
- HLA Laboratory, Beijing Red Cross Blood Center, Beijing, China
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University International Hospital, Beijing, China
| | - Song He Yang
- Faculty of Graduate Studies, Chengde Medical University, Chengde, Hebei, China
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei, China
| |
Collapse
|
16
|
Zeng Z, Fu Y, Li M, Shi Y, Ding Q, Chen S. Guben Qingfei decoction attenuates LPS-induced acute lung injury by modulating the TLR4/NF-κB and Keap1/Nrf2 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117674. [PMID: 38154525 DOI: 10.1016/j.jep.2023.117674] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 12/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury (ALI) is a life-threatening and widespread disease, with exceptionally high morbidity and mortality rates. Unfortunately, effective drugs for ALI treatment are currently lacking. Guben Qingfei decoction (GBQF) is a Chinese herbal compound known for its efficacy in treating viral pneumonia, yet the precise underlying mechanisms remain unknown. AIM OF THE STUDY This study aimed to validate the mitigating effect of GBQF on ALI and to further investigate its mechanism. MATERIALS AND METHODS An ALI mice model was established by infusing LPS into the endotracheal tube. The effects of GBQF on ALI were investigated by measuring lung W/D; MPO; BALF total protein concentration; total number of cells; TNF-α, IL-1β, and IL-6 levels; pathological changes in lung tissue, and oxidation products. Immunohistochemistry and Western Blotting were performed to verify the underlying mechanisms. MH-S and BEAS-2B cells were induced by LPS, and the effects of GBQF were confirmed by RT-PCR and immunofluorescence. RESULTS GBQF significantly reduced LPS-induced ALI in mice, improved lung inflammation, reduced the production of oxidative products, increased the activity of antioxidant enzymes, and reduced the degree of lung tissue damage. GBQF prevents MH-S cells from releasing inflammatory factors and reduces oxidative damage to BEAS-2B cells. In vivo studies have delved deeper into the mechanism of action of GBQF, revealing its correlation with the TLR4/NF-κB and Keap1/Nrf2 pathways. CONCLUSIONS Our study demonstrates that GBQF is an effective treatment for ALI, providing a new perspective on medication development for ALI treatment.
Collapse
Affiliation(s)
- Ziyuan Zeng
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Yuchen Fu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Minfang Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Qi Ding
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Sheng Chen
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.
| |
Collapse
|
17
|
Luo W, Song Z, Xu G, Wang H, Mu W, Wen J, Zhang P, Qin S, Xiao X, Bai Z. LicochalconeB inhibits cGAS-STING signaling pathway and prevents autoimmunity diseases. Int Immunopharmacol 2024; 128:111550. [PMID: 38232536 DOI: 10.1016/j.intimp.2024.111550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/19/2024]
Abstract
Cytosolic DNA activates the STING (stimulator of interferon genes) signaling pathway to trigger interferon and inflammatory responses that protect against microbial infections and cancer. However, Aicardi-Goutières syndrome (AGS) persistently activates the STING signaling pathway, which can lead to severe autoimmune diseases. We demonstrate herein that Licochalcone B (LicoB), the main component of traditional licorice, is an inhibitor of the STING signaling pathway. We observed that LicoB inhibited the activation of the STING signaling pathway in macrophages. Mechanically, LicoB affected the STING-TBK1-IRF3 signal axis and inhibited the activation of the STING downstream signaling pathway. Furthermore, LicoB inhibited the increase in type I interferon levels in mice induced by the STING agonist CMA. LicoB significantly reduced systemic inflammation in Trex1-/- mice. Our results show that LicoB, a STING signaling pathway inhibitor, is a promising candidate for the treatment of diseases related to STING signaling pathway activation.
Collapse
Affiliation(s)
- Wei Luo
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China; School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
| | - Zheng Song
- Peking University 302 Clinical Medical School, Beijing 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Guang Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Hongbo Wang
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Wenqing Mu
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Jincai Wen
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Ping Zhang
- Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing 100039, China
| | - Shuanglin Qin
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China; School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China.
| | - Xiaohe Xiao
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China.
| | - Zhaofang Bai
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China.
| |
Collapse
|