1
|
Li CL, Yeh KY, Huang WN, Yen CY, Wang KC, Liao EC, Chou TY, Huang HS, Yu SJ. Brevilin A inhibits IL‑17A‑induced inflammation in psoriasis by modulating HSP 70. Mol Med Rep 2025; 31:156. [PMID: 40211707 PMCID: PMC12001315 DOI: 10.3892/mmr.2025.13521] [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: 09/05/2024] [Accepted: 02/18/2025] [Indexed: 04/18/2025] Open
Abstract
Psoriasis, which is characterized by keratinocyte hyperproliferation, presents complex management challenges. The heat shock protein 70 (HSP 70) family, which is essential in protein folding and stress responses, also modulates inflammation, suggesting its therapeutic potential in inflammation‑driven diseases. The present study aimed to explore the effects of brevilin A, a natural compound known to alleviate imiquimod‑induced psoriasis, on HSP 70 expression and proinflammatory cytokine production in HaCaT cells stimulated with IL‑17A. An HSP 70 inhibitor was used to determine its role in cytokine regulation, and the effect of brevilin A on skin pathology in mice was examined via immunohistochemistry and hematoxylin and eosin staining. The results revealed that brevilin A markedly decreased IL‑6 and IL‑8 levels after IL‑17A stimulation at both 9 and 24 h in HaCaT cells, and increased HSP 70 and HSP 90 expression levels. Notably, the brevilin A‑induced suppression of cytokine levels was reversed when cells were co‑treated with the HSP 70 inhibitor. In vivo, brevilin A enhanced HSP 70 expression and reduced skin hyperproliferation. These findings suggested that brevilin A may modulate HSP 70 expression and dampen the inflammatory response induced by IL‑17A, indicating its potential as an innovative treatment for psoriasis.
Collapse
Affiliation(s)
- Chia-Ling Li
- Children's Medical Center, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
- Department of Physical Therapy, College of Medical and Health Care, Hungkuang University, Taichung 433, Taiwan, R.O.C
| | - Kuei-Ying Yeh
- Department of Physical Therapy, College of Medical and Health Care, Hungkuang University, Taichung 433, Taiwan, R.O.C
| | - Wen-Nan Huang
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan, R.O.C
- Department of Business Administration, Ling-Tung University, Taichung 408, Taiwan, R.O.C
- College of Medicine, National Chung Hsing University, Taichung 402, Taiwan, R.O.C
| | - Chung-Yang Yen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan, R.O.C
- Integrated Care Center of Psoriatic Disease, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
- Department of Dermatology, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
| | - Kai-Chun Wang
- Division of Allergy-Immunology-Rheumatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, R.O.C
- College of Medicine, National Sun Yat-sen University, Kaohsiung 804, Taiwan, R.O.C
| | - En-Chih Liao
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 252, Taiwan, R.O.C
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan, R.O.C
| | - Ting-Yu Chou
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
| | - Hung-Sen Huang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, R.O.C
| | - Sheng-Jie Yu
- Integrated Care Center of Psoriatic Disease, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
- Institute of Biomedical Sciences, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan, R.O.C
| |
Collapse
|
2
|
Singh V, Ubaid S, Kashif M, Singh T, Singh G, Pahwa R, Singh A. Role of inflammasomes in cancer immunity: mechanisms and therapeutic potential. J Exp Clin Cancer Res 2025; 44:109. [PMID: 40155968 PMCID: PMC11954315 DOI: 10.1186/s13046-025-03366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025] Open
Abstract
Inflammasomes are multi-protein complexes that detect pathogenic and damage-associated molecular patterns, activating caspase-1, pyroptosis, and the maturation of pro-inflammatory cytokines such as IL-1β and IL-18Within the tumor microenvironment, inflammasomes like NLRP3 play critical roles in cancer initiation, promotion, and progression. Their activation influences the crosstalk between innate and adaptive immunity by modulating immune cell recruitment, cytokine secretion, and T-cell differentiation. While inflammasomes can contribute to tumor growth and metastasis through chronic inflammation, their components also present novel therapeutic targets. Several inhibitors targeting inflammasome components- such as sensor proteins (e.g., NLRP3, AIM2), adaptor proteins (e.g., ASC), caspase-1, and downstream cytokines- are being explored to modulate inflammasome activity. These therapeutic strategies aim to modulate inflammasome activity to enhance anti-tumor immune responses and improve clinical outcomes. Understanding the role of inflammasomes in cancer immunity is crucial for developing interventions that effectively bridge innate and adaptive immune responses for better therapeutic outcomes.
Collapse
Affiliation(s)
- Vivek Singh
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Saba Ubaid
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Mohammad Kashif
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Tanvi Singh
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Gaurav Singh
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Roma Pahwa
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anand Singh
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
3
|
Kim JK, Sapkota A, Roh T, Jo EK. The intricate interactions between inflammasomes and bacterial pathogens: Roles, mechanisms, and therapeutic potentials. Pharmacol Ther 2025; 265:108756. [PMID: 39581503 DOI: 10.1016/j.pharmthera.2024.108756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/06/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Inflammasomes are intracellular multiprotein complexes that consist of a sensor, an adaptor, and a caspase enzyme to cleave interleukin (IL)-1β and IL-18 into their mature forms. In addition, caspase-1 and -11 activation results in the cleavage of gasdermin D to form pores, thereby inducing pyroptosis. Activation of the inflammasome and pyroptosis promotes host defense against pathogens, whereas dysregulation of the inflammasome can result in various pathologies. Inflammasomes exhibit versatile microbial signal detection, directly or indirectly, through cellular processes, such as ion fluctuations, reactive oxygen species generation, and the disruption of intracellular organelle function; however, bacteria have adaptive strategies to manipulate the inflammasome by altering microbe-associated molecular patterns, intercepting innate pathways with secreted effectors, and attenuating inflammatory and cell death responses. In this review, we summarize recent advances in the diverse roles of the inflammasome during bacterial infections and discuss how bacteria exploit inflammasome pathways to establish infections or persistence. In addition, we highlight the therapeutic potential of harnessing bacterial immune subversion strategies against acute and chronic bacterial infections. A more comprehensive understanding of the significance of inflammasomes in immunity and their intricate roles in the battle between bacterial pathogens and hosts will lead to the development of innovative strategies to address emerging threats posed by the expansion of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Asmita Sapkota
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
4
|
Wang R, Gao C, Yu M, Song J, Feng Z, Wang R, Pan H, Liu H, Li W, Fan X. Mechanistic prediction and validation of Brevilin A Therapeutic effects in Lung Cancer. BMC Complement Med Ther 2024; 24:214. [PMID: 38840248 PMCID: PMC11151568 DOI: 10.1186/s12906-024-04516-z] [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: 02/25/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Traditional Chinese medicine (TCM) has been found widespread application in neoplasm treatment, yielding promising therapeutic candidates. Previous studies have revealed the anti-cancer properties of Brevilin A, a naturally occurring sesquiterpene lactone derived from Centipeda minima (L.) A.Br. (C. minima), a TCM herb, specifically against lung cancer. However, the underlying mechanisms of its effects remain elusive. This study employs network pharmacology and experimental analyses to unravel the molecular mechanisms of Brevilin A in lung cancer. METHODS The Batman-TCM, Swiss Target Prediction, Pharmmapper, SuperPred, and BindingDB databases were screened to identify Brevilin A targets. Lung cancer-related targets were sourced from GEO, Genecards, OMIM, TTD, and Drugbank databases. Utilizing Cytoscape software, a protein-protein interaction (PPI) network was established. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene set enrichment analysis (GSEA), and gene-pathway correlation analysis were conducted using R software. To validate network pharmacology results, molecular docking, molecular dynamics simulations, and in vitro experiments were performed. RESULTS We identified 599 Brevilin A-associated targets and 3864 lung cancer-related targets, with 155 overlapping genes considered as candidate targets for Brevilin A against lung cancer. The PPI network highlighted STAT3, TNF, HIF1A, PTEN, ESR1, and MTOR as potential therapeutic targets. GO and KEGG analyses revealed 2893 enriched GO terms and 157 enriched KEGG pathways, including the PI3K-Akt signaling pathway, FoxO signaling pathway, and HIF-1 signaling pathway. GSEA demonstrated a close association between hub genes and lung cancer. Gene-pathway correlation analysis indicated significant associations between hub genes and the cellular response to hypoxia pathway. Molecular docking and dynamics simulations confirmed Brevilin A's interaction with PTEN and HIF1A, respectively. In vitro experiments demonstrated Brevilin A-induced dose- and time-dependent cell death in A549 cells. Notably, Brevilin A treatment significantly reduced HIF-1α mRNA expression while increasing PTEN mRNA levels. CONCLUSIONS This study demonstrates that Brevilin A exerts anti-cancer effects in treating lung cancer through a multi-target and multi-pathway manner, with the HIF pathway potentially being involved. These results lay a theoretical foundation for the prospective clinical application of Brevilin A.
Collapse
Affiliation(s)
- Ruixue Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Cuiyun Gao
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Meng Yu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jialing Song
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Zhenzhen Feng
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ruyu Wang
- School of clinical medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Haimeng Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Wei Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Xiangzhen Fan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
5
|
Liu X, Zang L, Yu J, Yu J, Wang S, Zhou L, Song H, Ma Y, Niu X, Li W. Anti-inflammatory effect of proanthocyanidins from blueberry through NF-κβ/NLRP3 signaling pathway in vivo and in vitro. Immunopharmacol Immunotoxicol 2024:1-11. [PMID: 38772618 DOI: 10.1080/08923973.2024.2358770] [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: 12/28/2023] [Accepted: 05/18/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Systemic inflammatory response syndrome (SIRS) is an uncontrolled systemic inflammatory response. Proanthocyanidins (PC) is a general term of polyphenol compounds widely existed in blueberry fruits and can treat inflammation-related diseases. This study aimed to explore the regulatory effect of PC on lipopolysaccharide (LPS)-induced systemic inflammation and its potential mechanism, providing effective strategies for the further development of PC. METHODS Here, RAW264.7 macrophages were stimulated with LPS to establish an inflammation model in vitro, while endotoxin shock mouse models were constructed by LPS in vivo. The function of PC was investigated by MTT, ELISA kits, H&E staining, immunohistochemistry, and Western blot analysis. RESULTS Functionally, PC could demonstrate the potential to mitigate mortality in mice with endotoxin shock, as well as attenuated the levels of inflammatory cytokines (IL-6, TNF-α) and biochemical indicators (AST, ALT, CRE and BUN). Moreover, it had a significant protective effect on lung and kidney tissues damage. Mechanistically, PC exerted anti-inflammatory effects by inhibiting the activation of the NF-κB/NLRP3 signaling pathway. CONCLUSION PC might have the potential ability of anti-inflammatory effects via modulation of the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Lulu Zang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yajing Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
6
|
Yang W, Wei Y, Sun J, Yao C, Ai F, Ding H. Safranal exerts a neuroprotective effect on Parkinson's disease with suppression of NLRP3 inflammation activation. Mol Biol Rep 2024; 51:593. [PMID: 38683404 PMCID: PMC11059006 DOI: 10.1007/s11033-024-09537-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/08/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a common central nervous system neurodegenerative disease. Neuroinflammation is one of the significant neuropathological hallmarks. As a traditional Chinese medicine, Safranal exerts anti-inflammatory effects in various diseases, however, whether it plays a similar effect on PD is still unclear. The study was to investigate the effects and mechanism of Safranal on PD. METHODS The PD mouse model was established by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine MPTP firstly. Next, the degree of muscle stiffness, neuromuscular function, motor retardation and motor coordination ability were examined by observing and testing mouse movement behavior. Immunofluorescence staining was used to observe the expression of tyrosine hydroxylase (TH). The dopamine (DA) content of the striatum was detected by High-performance liquid chromatography (HPLC). The expression of TH and NLRP3 inflammasome-related markers NLRP3, IL-1β, and Capase-1 were detected by Real-time Polymerase Chain Reaction (qRT-PCR) and western blotting (WB) respectively. RESULTS Through behavioral testing, Parkinson's mouse showed a higher muscle stiffness and neuromuscular tension, a more motor retardation and activity disorders, together with a worse motor coordination compared with sham group. Simultaneously, DA content and TH expression in the striatum were decreased. However, after using Safranal treatment, the above pathological symptoms of Parkinson's mouse all improved compared with Safranal untreated group, the DA content and TH expression were also increased to varying degrees. Surprisingly, it observed a suppression of NLRP3 inflammation in the striatum of Parkinson's mouse. CONCLUSIONS Safranal played a neuroprotective effect on the Parkinson's disease and its mechanism was related to the inhibition of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Wenping Yang
- Division of Neurology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, NO. 300 Guangzhou Road, Nanjing, China
| | - Yongyue Wei
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jin Sun
- Department of Nuclear Medicine, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Caixia Yao
- Departement of Endocrine, Nanjing Gao Chun People's Hospital, Nanjing, China
| | - Fen Ai
- Department of Emergency, Tongji Medical College, The Central Hospital of Wuhan, Huazhong University of Science and Technology, NO. 26 Shengli Street, Wuhan, Jiang'an District, China.
| | - Haixia Ding
- Division of Neurology, Department of Geriatrics, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, NO. 300 Guangzhou Road, Nanjing, China.
| |
Collapse
|
7
|
Yao J, Sterling K, Wang Z, Zhang Y, Song W. The role of inflammasomes in human diseases and their potential as therapeutic targets. Signal Transduct Target Ther 2024; 9:10. [PMID: 38177104 PMCID: PMC10766654 DOI: 10.1038/s41392-023-01687-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 09/18/2023] [Accepted: 10/13/2023] [Indexed: 01/06/2024] Open
Abstract
Inflammasomes are large protein complexes that play a major role in sensing inflammatory signals and triggering the innate immune response. Each inflammasome complex has three major components: an upstream sensor molecule that is connected to a downstream effector protein such as caspase-1 through the adapter protein ASC. Inflammasome formation typically occurs in response to infectious agents or cellular damage. The active inflammasome then triggers caspase-1 activation, followed by the secretion of pro-inflammatory cytokines and pyroptotic cell death. Aberrant inflammasome activation and activity contribute to the development of diabetes, cancer, and several cardiovascular and neurodegenerative disorders. As a result, recent research has increasingly focused on investigating the mechanisms that regulate inflammasome assembly and activation, as well as the potential of targeting inflammasomes to treat various diseases. Multiple clinical trials are currently underway to evaluate the therapeutic potential of several distinct inflammasome-targeting therapies. Therefore, understanding how different inflammasomes contribute to disease pathology may have significant implications for developing novel therapeutic strategies. In this article, we provide a summary of the biological and pathological roles of inflammasomes in health and disease. We also highlight key evidence that suggests targeting inflammasomes could be a novel strategy for developing new disease-modifying therapies that may be effective in several conditions.
Collapse
Affiliation(s)
- Jing Yao
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yun Zhang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Zhejiang Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
8
|
Liu YF, Li WQ, Hu ND, Ai B, Xia HX, Guo X, Chen Z, Xia H. Brevilin A ameliorates sepsis-induced cardiomyopathy through inhibiting NLRP3 inflammation. Ann Med Surg (Lond) 2023; 85:5952-5962. [PMID: 38098561 PMCID: PMC10718335 DOI: 10.1097/ms9.0000000000001403] [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: 08/22/2023] [Accepted: 10/06/2023] [Indexed: 12/17/2023] Open
Abstract
Background Sepsis is a systemic inflammatory disease, and Brevilin A (BA) has a powerful anti-inflammatory effect. However, whether BA has a similar effect on septic cardiomyopathy remains unclear. This study aimed to investigate the effect and mechanism of BA in septic cardiomyopathy. Methods First, a model of septic cardiomyopathy was constructed in vitro and in vivo. The expression of the cardiac injury markers, NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammation factors and its upstream modulator NF-κB was detected by real-time polymerase chain reaction and western blotting. Cardiac function was measured using echocardiography, cell viability was detected using the methyl thiazolyl tetrazolium assay. To further investigate the effects of BA on septic cardiomyopathy, different concentrations of BA were used. The experiment was divided into control group, LPS induced- group, LPS+2.5, 5.0, 10.0 μM BA treatment group of the vitro model, and the Sham, CLP, CLP+10, 20, 30 mg/kg BA treatment groups of the rat vivo model. Lastly, cardiac injury, NLRP3 inflammation, and cardiac function were assessed in each group. Results The mRNA and protein expression of cardiac inflammation and injury genes were significantly increased in the in vitro and in vivo sepsis cardiomyopathy models. When different concentrations of BA were used in sepsis cardiomyopathy in vivo and in vitro, the above-mentioned myocardial inflammation and injury factors were suppressed to varying degrees, cell viability increased, cardiac function improved, and the survival rate of rats also increased. Conclusion BA ameliorated sepsis cardiomyopathy by inhibiting NF-κB/NLRP3 inflammation activation.
Collapse
Affiliation(s)
- Ya-Feng Liu
- Department of Emergency, Renmin Hospital of Wuhan University
| | - Wen-Qiang Li
- Department of Emergency, Renmin Hospital of Wuhan University
| | - Nian-Dan Hu
- Department of Emergency, Renmin Hospital of Wuhan University
| | - Bo Ai
- Department of Emergency, Renmin Hospital of Wuhan University
| | - Hong-Xia Xia
- Department of Emergency, Renmin Hospital of Wuhan University
| | - Xin Guo
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Zheng Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, Hubei, People’s Republic of China
| |
Collapse
|
9
|
Ruan Q, Wang C, Zhang Y, Sun J. Brevilin A attenuates cartilage destruction in osteoarthritis mouse model by inhibiting inflammation and ferroptosis via SIRT1/Nrf2/GPX4 signaling pathway. Int Immunopharmacol 2023; 124:110924. [PMID: 37717314 DOI: 10.1016/j.intimp.2023.110924] [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: 08/18/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
Osteoarthritis (OA) is a serious orthopedic disease that affects people's quality of life. Although there are many treatment methods, the treatment effect is still not good. Brevilin A is a bioactive compound isolated from the medicinal herbCentipeda minima. The potential efficacy of brevilin A on OA was explored in this study. Mouse chondrocytes were isolated and stimulated by IL-1β and mouse OA model was induced by destabilization of the medial meniscus (DMM). The results demonstrated that brevilin A markedly inhibited IL-1β-induced MMP1 and MMP3 production. IL-1β-induced PGE2, NO, MDA, and iron production were alleviated by brevilin A. The production of GSH and the expression of SIRT1, Nrf2, HO-1, GPX4, and Ferritin were increased by brevilin A. Furthermore, the inhibition of brevilin A on IL-1β-induced inflammation and ferroptosis were prevented by SIRT1 inhibitor. In vivo, the results showed brevilin A markedly attenuated OA progression in DMM-induced mouse OA model. Also, brevilin A could alleviate MMP1, MMP3, iNOS, and COX2 expression in OA mice. In conclusion, brevilin A protected mice against OA via suppressing inflammatory response and ferroptosis by regulating SIRT1/Nrf2/GPX4 signaling.
Collapse
Affiliation(s)
- Qing Ruan
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Cuijie Wang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China
| | - Yunfeng Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Jiayang Sun
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China.
| |
Collapse
|
10
|
Wei J, Lin Z, Dai Z, Zhou Z, Bi Y, Zheng R, Hu X, Xu Z, Yuan G, Wang W. Brevilin A inhibits RANKL-induced osteoclast differentiation and bone resorption. In Vitro Cell Dev Biol Anim 2023; 59:420-430. [PMID: 37460875 DOI: 10.1007/s11626-023-00783-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/20/2023] [Indexed: 07/29/2023]
Abstract
Brevilin A (BA) is the primary component of Centipeda minima, which is widely used in Chinese traditional medicine. The anti-inflammatory and anti-tumor properties of BA have been established; however, its function in bone metabolism is not well understood. This study revealed that concentrations of BA below 1.0 µM did not inhibit the proliferation of bone marrow macrophages but did impede the differentiation and bone resorption activity of osteoclasts. Furthermore, BA suppressed the expression of osteoclast-specific genes Mmp9, Acp5, Dc-stamp, Ctsk, and Atp6v0d2. In addition, mTOR, ERK, and NFATc1 activation in bone marrow macrophages were suppressed by BA. As a whole, BA blocks the mTOR and ERK signaling pathways, which is responsible for the development and activity of osteoclasts, and the resorption of bone.
Collapse
Affiliation(s)
- Jinfu Wei
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Zihong Lin
- Department of Shantou Central Hospital, Shantou, 515000, Guangdong, China
| | - Zeyu Dai
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Zibin Zhou
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Yonghao Bi
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Ruiwu Zheng
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Xianghua Hu
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Zhaoxin Xu
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Guixin Yuan
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, 515000, Guangdong, China.
| | - Weidong Wang
- Department of Bone and Soft Tissue Oncology Surgery, Cancer Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China.
| |
Collapse
|
11
|
Gao C, Pan H, Ma F, Zhang Z, Zhao Z, Song J, Li W, Fan X. Centipeda minima active components and mechanisms in lung cancer. BMC Complement Med Ther 2023; 23:89. [PMID: 36959600 PMCID: PMC10035269 DOI: 10.1186/s12906-023-03915-y] [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: 10/31/2022] [Accepted: 03/09/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Traditional Chinese medicine (TCM) has been extensively used for neoplasm treatment and has provided many promising therapeutic candidates. We previously found that Centipeda minima (C. minima), a Chinese medicinal herb, showed anti-cancer effects in lung cancer. However, the active components and underlying mechanisms remain unclear. In this study, we used network pharmacology to evaluate C. minima active compounds and molecular mechanisms in lung cancer. METHODS We screened the TCMSP database for bioactive compounds and their corresponding potential targets. Lung cancer-associated targets were collected from Genecards, OMIM, and Drugbank databases. We then established a drug-ingredients-gene symbols-disease (D-I-G-D) network and a protein-protein interaction (PPI) network using Cytoscape software, and we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses using R software. To verify the network pharmacology results, we then performed survival analysis, molecular docking analysis, as well as in vitro and in vivo experiments. RESULTS We identified a total of 21 C. minima bioactive compounds and 179 corresponding targets. We screened 804 targets related to lung cancer, 60 of which overlapped with C. minima. The top three candidate ingredients identified by D-I-G-D network analysis were quercetin, nobiletin, and beta-sitosterol. PPI network and core target analyses suggested that TP53, AKT1, and MYC are potential therapeutic targets. Moreover, molecular docking analysis confirmed that quercetin, nobiletin, and beta-sitosterol, combined well with TP53, AKT1, and MYC respectively. In vitro experiments verified that quercetin induced non-small cell lung cancer (NSCLC) cell death in a dose-dependent manner. GO and KEGG analyses found 1771 enriched GO terms and 144 enriched KEGG pathways, including a variety of cancer related pathways, the IL-17 signaling pathway, the platinum drug resistance pathway, and apoptosis pathways. Our in vivo experimental results confirmed that a C. minima ethanol extract (ECM) enhanced cisplatin (CDDP) induced cell apoptosis in NSCLC xenografts. CONCLUSIONS This study revealed the key C. minima active ingredients and molecular mechanisms in the treatment of lung cancer, providing a molecular basis for further C. minima therapeutic investigation.
Collapse
Affiliation(s)
- Cuiyun Gao
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fengjun Ma
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ze Zhang
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Zedan Zhao
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Jialing Song
- School of Rehabilitation Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Wei Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Xiangzhen Fan
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
12
|
Conceição M, Beserra FP, Aldana Mejia JA, Caldas GR, Tanimoto MH, Luzenti AM, Gaspari PDM, Evans ND, Bastos JK, Pellizzon CH. Guttiferones: An insight into occurrence, biosynthesis, and their broad spectrum of pharmacological activities. Chem Biol Interact 2023; 370:110313. [PMID: 36566914 DOI: 10.1016/j.cbi.2022.110313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
Guttiferones belong to the polyisoprenylated benzophenone, a class of compounds, a very restricted group of natural plant products, especially in the Clusiaceae family. They are commonly found in bark, stem, leaves, and fruits of plants of the genus Garcinia and Symphonia. Guttiferones have the following classifications according to their chemical structure: A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, P, Q, R, S, and T. All of them have received growing attention due to its multiple biological activities. This review provides a first comprehensive approach to plant sources, phytochemical profile, specific pharmacological effects, and mechanisms of guttiferones already described. Studies indicate a broad spectrum of pharmacological activities, such as: anti-inflammatory, immunomodulatory, antioxidant, antitumor, antiparasitic, antiviral, and antimicrobial. Despite the low toxicity of these compounds in healthy cells, there is a lack of studies in the literature related to toxicity in general. Given their beneficial effects, guttiferones are expected to be great potential drug candidates for treating cancer and infectious and transmissible diseases. However, further studies are needed to elucidate their toxicity, specific molecular mechanisms and targets, and to perform more in-depth pharmacokinetic studies. This review highlights chemical properties, biological characteristics, and mechanisms of action so far, offering a broad view of the subject and perspectives for the future of guttiferones in therapeutics.
Collapse
Affiliation(s)
- Mariana Conceição
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Fernando Pereira Beserra
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil; Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.
| | - Jennyfer Andrea Aldana Mejia
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Gabriel Rocha Caldas
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Matheus Hikaru Tanimoto
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Andréia Marincek Luzenti
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Priscyla Daniely Marcato Gaspari
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Nicholas David Evans
- Human Development and Health, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Jairo Kenupp Bastos
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Cláudia Helena Pellizzon
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
13
|
Liu Y, Wang D, Li T, Yang F, Li Z, Bai X, Wang Y. The role of NLRP3 inflammasome in inflammation-related skeletal muscle atrophy. Front Immunol 2022; 13:1035709. [PMID: 36405697 PMCID: PMC9668849 DOI: 10.3389/fimmu.2022.1035709] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/13/2022] [Indexed: 04/04/2024] Open
Abstract
Skeletal muscle atrophy is a common complication in survivors of sepsis, which affects the respiratory and motor functions of patients, thus severely impacting their quality of life and long-term survival. Although several advances have been made in investigations on the pathogenetic mechanism of sepsis-induced skeletal muscle atrophy, the underlying mechanisms remain unclear. Findings from recent studies suggest that the nucleotide-binding and oligomerisation domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, a regulator of inflammation, may be crucial in the development of skeletal muscle atrophy. NLRP3 inhibitors contribute to the inhibition of catabolic processes, skeletal muscle atrophy and cachexia-induced inflammation. Here, we review the mechanisms by which NLRP3 mediates these responses and analyse how NLRP3 affects muscle wasting during inflammation.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongfang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Yang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Activation and Pharmacological Regulation of Inflammasomes. Biomolecules 2022; 12:biom12071005. [PMID: 35883561 PMCID: PMC9313256 DOI: 10.3390/biom12071005] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023] Open
Abstract
Inflammasomes are intracellular signaling complexes of the innate immune system, which is part of the response to exogenous pathogens or physiological aberration. The multiprotein complexes mainly consist of sensor proteins, adaptors, and pro-caspase-1. The assembly of the inflammasome upon extracellular and intracellular cues drives the activation of caspase-1, which processes pro-inflammatory cytokines IL-1β and IL-18 to maturation and gasdermin-D for pore formation, leading to pyroptosis and cytokine release. Inflammasome signaling functions in numerous infectious or sterile inflammatory diseases, including inherited autoinflammatory diseases, metabolic disorders, cardiovascular diseases, cancers, neurodegenerative disorders, and COVID-19. In this review, we summarized current ideas on the organization and activation of inflammasomes, with details on the molecular mechanisms, regulations, and interventions. The recent developments of pharmacological strategies targeting inflammasomes as disease therapeutics were also covered.
Collapse
|
15
|
Tan J, Qiao Z, Meng M, Zhang F, Kwan HY, Zhong K, Yang C, Wang Y, Zhang M, Liu Z, Su T. Centipeda minima: An update on its phytochemistry, pharmacology and safety. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115027. [PMID: 35091011 DOI: 10.1016/j.jep.2022.115027] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/07/2022] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Centipeda minima (CM), the dried whole plant of Centipeda minima (L.) A. Braun and Aschers, has been used as a traditional Chinese medicinal herb for thousands of years for the treatments of rhinitis, sinusitis, cough and asthmatic diseases. This review aimed to evaluate the therapeutic potential of CM by summarizing its phytochemistry, pharmacology, clinical application and safety. METHODS This review summarizes the published studies on CM in the Chinese Pharmacopoeia and literature databases including PubMed, Web of Science, Baidu Scholar, Wiley and China Knowledge Resource Integrated Database (CNKI), as well as the research articles on the phytochemistry, pharmacology, clinical application and safety of CM. RESULTS A total of 191 compounds have been isolated and identified from CM, including terpenes, flavonoids, sterols, phenols, organic acids and volatile oils. In addition, the pharmacological effects of CM, such as anti-cancer, anti-inflammatory and anti-bacterial activities, have also been evaluated by both in vitro and in vivo studies. The signaling pathways and mechanisms of action underlying the anti-cancer effects of CM have been revealed. Clinical applications of CM mainly include rhinitis and sinusitis, gynecological inflammation, cough, as well as asthma. CONCLUSION CM is a medicinal herb that possesses many therapeutic effects. Cutting-edge technology and system biology could provide us a more comprehensive understanding of the therapeutic effects, constituting components and toxicity of CM, which are the prerequisites for its translation into therapeutics for various disease treatments.
Collapse
Affiliation(s)
- Jincheng Tan
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Zhiping Qiao
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Mingjing Meng
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Fan Zhang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Hiu Yee Kwan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Keying Zhong
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Chunfang Yang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Yechun Wang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Mi Zhang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Tao Su
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau.
| |
Collapse
|
16
|
Lee D, Kwak HJ, Kim BH, Kim DW, Kim HY, Kim SH, Kang KS. Brevilin A Isolated from Centipeda minima Induces Apoptosis in Human Gastric Cancer Cells via an Extrinsic Apoptotic Signaling Pathway. PLANTS 2022; 11:plants11131658. [PMID: 35807611 PMCID: PMC9268799 DOI: 10.3390/plants11131658] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022]
Abstract
Brevilin A, which has anticancer activities against a range of cancers, is an abundant constituent of the medicinal herb Centipeda minima (L.) A. Braun & Asch, which has also been reported to have anticancer activity against breast cancer cells. However, the anticancer activities of C. minima and brevilin A against human gastric cancer have yet to be reported. In this study, we aimed to evaluate the cytotoxicity and molecular basis underlying the anticancer activities of extracts of C. minima (CMX) and brevilin A against human gastric cancer (AGS) cells. We deduced the potential targets and mechanisms underlying the anticancer activity of brevilin A based on a network pharmacology approach. CCND1, CDK4, and BCL2L1 were identified as the key anticancer genes targeted by brevilin A. Cytotoxicity analyses revealed that CMX and brevilin A reduced the viability of AGS cells to levels below 50% (9.73 ± 1.29 µg/mL and 54.69 ± 1.38 μM, respectively). Furthermore, Hoechst 33342, annexin V, and propidium iodide staining and western blot analyses revealed that CMX and brevilin A promoted a significant induction of apoptotic cell death by upregulating the expression of cleaved caspase-8 and cleaved caspase-3 and reducing the ratio of Bax to Bcl-2, which is partially consistent with the findings of our network pharmacology analysis. Collectively, our observations indicate that CMX and brevilin A are novel sources of herbal medicine with potential utility as effective agents for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
| | - Hee Jae Kwak
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea;
| | | | - Dong-Wook Kim
- College of Pharmacy, Wonkwang University, Iksan 54538, Korea;
| | - Hyun Young Kim
- Department of Food Science and Nutrition, Gyeongsang National University, Jinju 52725, Korea;
| | - Seung Hyun Kim
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea;
- Correspondence: (S.H.K.); (K.S.K.); Tel.: +82-32-749-4514 (S.H.K.); +82-31-750-5402 (K.S.K.)
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
- Correspondence: (S.H.K.); (K.S.K.); Tel.: +82-32-749-4514 (S.H.K.); +82-31-750-5402 (K.S.K.)
| |
Collapse
|
17
|
Yeshi K, Turpin G, Jamtsho T, Wangchuk P. Indigenous Uses, Phytochemical Analysis, and Anti-Inflammatory Properties of Australian Tropical Medicinal Plants. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123849. [PMID: 35744969 PMCID: PMC9231311 DOI: 10.3390/molecules27123849] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 11/16/2022]
Abstract
Australian tropical plants have been a rich source of food (bush food) and medicine to the first Australians (Aboriginal people), who are believed to have lived for more than 50,000 years. Plants such as spreading sneezeweed (Centipeda minima), goat’s foot (Ipomoea pes-caprae), and hop bush (Dodonaea viscosa and D. polyandra) are a few popular Aboriginal medicinal plants. Thus far, more than 900 medicinal plants have been recorded in the tropical region alone, and many of them are associated with diverse ethnomedicinal uses that belong to the traditional owners of Aboriginal people. In our effort to find anti-inflammatory lead compounds in collaboration with Aboriginal communities from their medicinal plants, we reviewed 78 medicinal plants used against various inflammation and inflammatory-related conditions by Aboriginal people. Out of those 78 species, we have included only 45 species whose crude extracts or isolated pure compounds showed anti-inflammatory properties. Upon investigating compounds isolated from 40 species (for five species, only crude extracts were studied), 83 compounds were associated with various anti-inflammatory properties. Alphitolic acid, Betulinic acid, Malabaric acid, and Hispidulin reduced proinflammatory cytokines and cyclooxygenase enzymes (COX-1 and 2) with IC50 values ranging from 11.5 to 46.9 uM. Other promising anti-inflammatory compounds are Brevilin A (from Centipeda minima), Eupalestin, and 5′-methoxy nobiletin (from Ageratum conyzoides), Calophyllolide (from Calophyllum inophyllum), and Brusatol (from Brucea javanica). D. polyandra is one example of an Aboriginal medicinal plant from which a novel anti-inflammatory benzoyl ester clerodane diterpenoid compound was obtained (compound name not disclosed), and it is in the development of topical medicines for inflammatory skin diseases. Medicinal plants in the tropics and those associated with indigenous knowledge of Aboriginal people could be a potential alternative source of novel anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Karma Yeshi
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
- Correspondence:
| | - Gerry Turpin
- Tropical Herbarium of Australia, James Cook University, Building E1, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
| | - Tenzin Jamtsho
- Yangchenphug High School, Ministry of Education, Thimphu 11001, Bhutan;
| | - Phurpa Wangchuk
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
| |
Collapse
|
18
|
Liu L, Chen X, Jiang Y, Yuan Y, Yang L, Hu Q, Tang J, Meng X, Xie C, Shen X. Brevilin A Ameliorates Acute Lung Injury and Inflammation Through Inhibition of NF-κB Signaling via Targeting IKKα/β. Front Pharmacol 2022; 13:911157. [PMID: 35774606 PMCID: PMC9237443 DOI: 10.3389/fphar.2022.911157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Acute lung injury (ALI) is life-threatening disease characterized by uncontrolled inflammatory response. IKKα/β, the key kinases in the activation of NF-κB pathway, are implicated in inflammatory pulmonary injury, and represent attractive targets for ALI therapy. Brevilin A (BVA) is a sesquiterpene lactone from Centipeda minima, a Chinese herb used to treat inflammatory diseases. This study aims to investigate the inhibition of BVA on ALI, with focus on clarifying the molecular mechanisms involved in BVA-mediated anti-inflammatory activity in macrophages. Briefly, BVA significantly inhibited the production of NO and PGE2 by suppressing iNOS and COX2 expression, and suppressed the mRNA expression of IL-1β, IL-6, and TNFα in LPS/IFNγ-stimulated RAW264.7 macrophages. The anti-inflammatory activity of BVA was further confirmed in LPS/IFNγ-stimulated BMDMs and TNFα/IFNγ-exposed RAW264.7 cells. In vivo, BVA effectively attenuated LPS-induced lung damage, inflammatory infiltration, and production of pro-inflammatory cytokines, including MPO, IL-1β, IL-6, TNFα, and PGE2. Mechanistically, BVA could covalently bind to the cysteine 114 of IKKα/β, and effectively inhibiting the activity and function of IKKα/β, thereby resulting in the suppression of phosphorylation and degradation of IκBα and the subsequent activation of NF-κB signaling. Furthermore, pretreatment of DTT, a thiol ligand donor, significantly abolished BVA-mediated effects in LPS/IFNγ-stimulated RAW264.7 cells, suggesting the crucial role of the electrophilic α, β-unsaturated ketone of BVA on its anti-inflammatory activity. These results suggest that BVA ameliorates ALI through inhibition of NF-κB signaling via covalently targeting IKKα/β, raising the possibility that BVA could be effective in the treatment of ALI and other diseases harboring aberrant NF-κB signaling.
Collapse
Affiliation(s)
- Lu Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xian Chen
- Department of Pathology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yifang Jiang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun Yuan
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luyao Yang
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiongying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunguang Xie
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Chunguang Xie, ; Xiaofei Shen,
| | - Xiaofei Shen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Chunguang Xie, ; Xiaofei Shen,
| |
Collapse
|