1
|
Zhang Y, Wang L, Zeng J, Shen W. Research advances in polyphenols from Chinese herbal medicine for the prevention and treatment of chronic obstructive pulmonary disease: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03945-y. [PMID: 40035820 DOI: 10.1007/s00210-025-03945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is a global health problem due to its high death and morbidity worldwide, which is characterized by an incompletely reversible limitation in airflow that is not fully reversible. Unfortunately, Western medical treatments are unable to reverse the progressive decline in lung function. Importantly, polyphenolic compounds isolated from Chinese herbal medicine exhibited therapeutic/interventional effects on COPD in preclinical studies. This review systematically analyzed the pathogenesis of COPD, such as inflammation, oxidative stress, protease/antiprotease imbalance, aging, cell death, and dysbiosis of gut microbiota. Moreover, this review summarized the regulatory mechanisms of natural polyphenolic compounds for the treatment of COPD. Several studies have demonstrated that natural polyphenolic compounds have therapeutic effects on COPD by regulating various biological processes, such as anti-inflammatory, reduction of oxidative damage, anti-cell death, and inhibition of airway hyperglycemia. Mechanistically, this review found that the promising effects of natural polyphenolic compounds on COPD were mainly achieved through modulating the NF-κB and MAPK inflammatory pathways, the Nrf2 oxidative stress pathway, and the SIRT1/PGC-1α lung injury pathway. Furthermore, this review analyzed the efficacy and safety of natural polyphenolic compounds for the treatment of COPD in clinical trials, and discussed their challenges and future development directions. In conclusion, this review combined the latest literature to illustrate the various pathogenesis and interrelationships of COPD in the form of graphs, texts, and tables, and sorted out the functional role and mechanisms of natural polyphenols in treating COPD, with a view to providing new ideas and plans for the in-depth research on COPD and the systemic treatment of COPD with Chinese herbal medicine.
Collapse
Affiliation(s)
- Yang Zhang
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China
| | - Lijuan Wang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Jinyi Zeng
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China
| | - Wen Shen
- Department of General Practice Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Avenue, Wuhua District, Kunming, 650101, China.
| |
Collapse
|
2
|
Wang M, Hao Y, He W, Jia H, Zhong Z, Xia S. Nebulized mesenchymal stem cell-derived exosomes attenuate airway inflammation in a rat model of chronic obstructive pulmonary disease. Cell Immunol 2025; 409-410:104933. [PMID: 40020434 DOI: 10.1016/j.cellimm.2025.104933] [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: 11/25/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is one of the leading causes of death worldwide, and current treatments fail to significantly halt its progression. Exosomes derived from mesenchymal stem cells (MSCs-Exos) have demonstrated promising potential in treating COPD due to their anti-inflammatory and regenerative biological properties. In this study, we investigated the potential anti-inflammatory effects of bone marrow mesenchymal stem cell-derived exosomes (BMSCs-Exos) in a COPD rat model and the possible mechanisms by which they inhibit airway remodeling, as well as identifying the optimal dosage and administration route. Our results show that nebulized BMSC-Exos significantly improve lung function in COPD rats while reducing pulmonary inflammatory infiltration, bronchial mucus secretion, and collagen deposition. Moreover, BMSC-Exos treatment notably decreased the expression of pro-inflammatory cytokines such as TNF-α, IL-6 and IL-1β, and the pro-fibrotic factor TGF-β1 in serum, bronchoalveolar lavage fluid (BALF), and lung tissue. The most pronounced therapeutic effect was observed at a low dose of exosomes. Furthermore, quantitative real-time PCR and immunohistochemical analyses revealed that nebulized BMSC-Exos significantly inhibited airway remodeling and epithelial-mesenchymal transition (EMT) by suppressing the Wnt/β-catenin signaling pathway. In conclusion, these findings indicate that nebulized BMSC-Exos offer a noninvasive therapeutic strategy for COPD by mitigating lung inflammation and airway remodeling through the suppression of abnormal Wnt/β-catenin pathway activation induced by cigarette smoke (CS) and lipopolysaccharide (LPS) in rats.
Collapse
Affiliation(s)
- Min Wang
- Graduate School of Dalian Medical University, Dalian 116044, China
| | - Yuxin Hao
- Graduate School of Shandong First Medical University, Jinan 271016, China
| | - Wei He
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China
| | - Hui Jia
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China
| | - Zhaoshuang Zhong
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China
| | - Shuyue Xia
- Graduate School of Dalian Medical University, Dalian 116044, China; Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China.
| |
Collapse
|
3
|
Ji J, Ding X, Liu C, Dai L, Yu J, Li L, Wan S, Song Y, Zhao J, Su Z, Jia K, Li S. Efficacy and safety of anemoside B4 in canine pneumonia treatment: a prospective, randomized controlled trial. Front Vet Sci 2025; 12:1530318. [PMID: 40012747 PMCID: PMC11861352 DOI: 10.3389/fvets.2025.1530318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
Background Canine pneumonia is a serious respiratory disease often associated with Canine Infectious Respiratory Disease (CIRD). Current treatment strategies primarily rely on antibiotics and corticosteroids; however, the emergence of antibiotic resistance and potential side effects from prolonged corticosteroid use limit the effectiveness of these therapies in clinical practice. These challenges highlight the urgent need for alternative treatments. Anemoside B4 (AB4), derived from the traditional Chinese medicine Pulsatilla, has shown promise in preclinical studies for modulating inflammatory responses and improving clinical symptoms of pneumonia. Therefore, AB4 may offer a valuable alternative treatment option for canine pneumonia in veterinary medicine. Methods A prospective, randomized controlled trial was conducted at the Veterinary Drug Research and Evaluation Center of South China Agricultural University. Seventy-two dogs with mild-to-moderate pneumonia were randomly assigned to one of three groups: AB4, placebo, or Chuanxinlian injection (CXL). The primary outcome was the effect of AB4 on comprehensive clinical scoring of canine pneumonia; secondary outcomes included recovery times for primary symptoms and efficacy assessments. Additionally, AB4's safety in clinical applications was evaluated. Results The AB4 group demonstrated significantly lower composite clinical scores on Days 7 and 14 compared to the placebo group (p = 0.033 and p = 0.000, respectively). Significant differences in recovery times for fever and dyspnea were observed between the AB4 and placebo groups (p = 0.041 and p = 0.024, respectively). Moreover, the cure rate and overall efficacy on Day 14 were significantly higher in the AB4 group than those in the placebo group (p = 0.001 and p = 0.009, respectively). Conclusion These findings suggest that AB4 may be a promising treatment option for canine pneumonia, potentially serving as an alternative to traditional therapies. Further research is needed to explore its clinical potential in veterinary medicine.
Collapse
Affiliation(s)
- Jinzhao Ji
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Xiaoqing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Chuanli Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Lingling Dai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Junting Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Linghao Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Shaobing Wan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Yangyang Song
- Guangxi Innovate Pharmaceutical Co., Ltd, Guangxi, China
| | - Junqing Zhao
- Guangxi Innovate Pharmaceutical Co., Ltd, Guangxi, China
| | - Zhetong Su
- Guangxi Innovate Pharmaceutical Co., Ltd, Guangxi, China
| | - Kun Jia
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Shoujun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| |
Collapse
|
4
|
Duan W, Zhou Z, Huang Y, Cui Y, Jin X, Liu R, Chen L. Euphorbia helioscopia L. inhibits lung tumorigenesis through alleviating exhausted T cell induced by chronic inflammation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119097. [PMID: 39537116 DOI: 10.1016/j.jep.2024.119097] [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: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euphorbia helioscopia L. (ZQ) is a very effective traditional Chinese medicine for treating pneumonia and lung cancer. However, the effects and mechanisms by which ZQ prevents lung tumorigenesis in the presence of chronic inflammation remain unexplored. AIM To examine the effects and mechanisms of ZQ in alleviating chronic inflammation-induced T cell exhaustion and inhibiting lung tumorigenesis. METHODS A mice model of lung tumorigenesis under chronic inflammation conditions was established by repeated administration of lipopolysaccharide (LPS) and exposure to the tobacco carcinogen nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Mice were treated with ZQ (0.9, 1.8, and 3.6 g/kg/day) for 25 weeks. Lung pathology and tumor incidence were assessed, and inflammatory cytokine levels in bronchoalveolar lavage fluid (BALF) and serum were measured. Additionally, the proportions of CD3+ T, CD4+ T, and CD8+ T cells and their inhibitory receptors expression were evaluated. In vitro, T cell exhaustion models were induced using inflammatory-conditioned media, followed by treatment with ZQ (0.5, 2, 8 μg/mL). T cell exhaustion markers and characteristics were analyzed, and molecular mechanisms were explored using RNA sequencing and Immunoblotting analysis. RESULTS In vivo, ZQ significantly reduced inflammatory infiltration and lung damage, tumor incidence, number, size, and lung and spleen indices in mice. It also markedly lowered the levels of pro-inflammatory cytokines and immunosuppressive cytokines in BALF and serum. Additionally, ZQ improved the proportions of CD3+ T, CD4+ T, and CD8+ T cells and significantly decreased the expression of inhibitory receptors on CD4+ T and CD8+ T cells in the lung tissues and spleen. In vitro, ZQ effectively alleviated T cell exhaustion induced by the inflammatory environment, marked by reduced expression of inhibitory receptors, increased cytokine secretion, improved proliferation, and enhanced tumoricidal activity. RNA sequencing revealed that ZQ significantly downregulated the JAK-STAT signaling and upregulated stemness-associated pathways. Immunoblotting results indicated that treatment with ZQ markedly reduced the phosphorylation of Signal transducer and activator of transcription 3 (STAT3) and increased the expression of T cell factor -1/7 (TCF1/7). CONCLUSION ZQ inhibits lung tumorigenesis in LPS/NNK-treated mice through alleviating exhausted T cells induced by chronic inflammation, which is attributed to the suppression of STAT3 activation and the maintenance of stemness characteristics in T cells. These findings provide experimental evidence for the potential use of ZQ in preventing and treating lung tumourigenesis in patients with chronic inflammation and the clinical management of lung cancer patients with concomitant chronic inflammation.
Collapse
Affiliation(s)
- Wenbin Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Ziye Zhou
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yuqing Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yaru Cui
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Xuhui Jin
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Ronghua Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Lanying Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| |
Collapse
|
5
|
Luan X, Zhu D, Hao Y, Xie J, Wang X, Li Y, Zhu J. Qibai Pingfei Capsule ameliorated inflammation in chronic obstructive pulmonary disease (COPD) via HIF-1 α/glycolysis pathway mediated of BMAL1. Int Immunopharmacol 2025; 144:113636. [PMID: 39579541 DOI: 10.1016/j.intimp.2024.113636] [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: 08/14/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is characterized for the persistent inflammation. The brain and muscle arnt-like 1 (BMAL1), as a crucial clock gene, is associated with the expression level of upstream factor hypoxia-inducible factor (HIF)-1α in glycolysis, which may affect the occurrence of inflammatory reactions in COPD. However, the moderation effect of Qibai Pingfei Capsule (QBPF) capsule is still unknown on BMAL1 and HIF-1α/glycolytic pathway. OBJECTIVE The aim of this study is to investigate the modulatory effects of QBPF capsules on BMAL1 and the HIF-1α/glycolytic pathway in COPD. METHODS The multifactorial approach were used to construct the COPD rat model, including forced swimming, hypoxia, and inhalation of cigarette smoke with four weeks. Nextly, the rats received a two-week course of gavage treatment with medicant. Finally, tissue samples were collected for comprehensive analysis using various molecular biology techniques. These methods included molecular docking, immunoprecipitation, small interfering RNA (siRNA), hematoxylin and eosin (HE) staining, western blot (WB), and immunofluorescence etc. to elucidate the modulatory effects of QBPF for treating COPD in vitro and in vivo. RESULTS The expression levels in mRNA and protein of BMAL1 decreased in COPD, while the content in mRNA and protein of HIF-1α increased. At the same time, the concentration in glycolytic metabolites of hexokinase (HK), phosphofructokinase (PFK), pyruvate kinase (PK), lactate dehydrogenase (LDH), and lactate (LD) increased, and ATP decreased. The QBPF capsule can reverse the imbalance between BMAL1 and HIF-1α, improve disorders of glycolytic pathway, and alleviate the inflammation response. Notably, in vivo experiments, the interaction between BMAL1 and HIF-1α were confirmed via molecular docking and immunoprecipitation. In rescue experiments, intervention with siRNA BMAL1 in 16HBE cells revealed a significant decrease in BMAL1 levels and the therapeutic effect of QBPF was also affected. CONCLUSION QBPF could up-regulate the expression level of clock gene BMAL1, thereby regulating the HIF-1α/glycolytic pathway and metabolite to improve the inflammatory response in COPD.
Collapse
Affiliation(s)
- Xuejing Luan
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Dandan Zhu
- Research Center of Integrated Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu 241002, China
| | - Yifei Hao
- Research Center of Integrated Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu 241002, China
| | - Jinghui Xie
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xiu Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu 241002, China
| | - Yan Li
- Yijishan Hospital Affiliated to Wannan Medical College, Wuhu 241001, China
| | - Jie Zhu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; Research Center of Integrated Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu 241002, China.
| |
Collapse
|
6
|
Hu Y, Fan Q, Qiao B, Xu O, Lv B, Han N, Zhang X. Alleviatory Role of Panax Notoginseng Saponins in Modulating Inflammation and Pulmonary Vascular Remodeling in Chronic Obstructive Pulmonary Disease: mechanisms and Implications. COPD 2024; 21:2329282. [PMID: 38622983 DOI: 10.1080/15412555.2024.2329282] [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: 10/17/2023] [Accepted: 03/06/2024] [Indexed: 04/17/2024]
Abstract
COPD is an inflammatory lung disease that limits airflow and remodels the pulmonary vascular system. This study delves into the therapeutic potential and mechanistic underpinnings of Panax notoginseng Saponins (PNS) in alleviating inflammation and pulmonary vascular remodeling in a COPD rat model. Symmap and ETCM databases provided Panax notoginseng-related target genes, and the CTD and DisGeNET databases provided COPD-related genes. Intersection genes were subjected to protein-protein interaction analysis and pathway enrichment to identify downstream pathways. A COPD rat model was established, with groups receiving varying doses of PNS and a Roxithromycin control. The pathological changes in lung tissue and vasculature were examined using histological staining, while molecular alterations were explored through ELISA, RT-PCR, and Western blot. Network pharmacology research suggested PNS may affect the TLR4/NF-κB pathway linked to COPD development. The study revealed that, in contrast to the control group, the COPD model exhibited a significant increase in inflammatory markers and pathway components such as TLR4, NF-κB, HIF-1α, VEGF, ICAM-1, SELE mRNA, and serum TNF-α, IL-8, and IL-1β. Treatment with PNS notably decreased these markers and mitigated inflammation around the bronchi and vessels. Taken together, the study underscores the potential of PNS in reducing lung inflammation and vascular remodeling in COPD rats, primarily via modulation of the TLR4/NF-κB/HIF-1α/VEGF pathway. This research offers valuable insights for developing new therapeutic strategies for managing and preventing COPD.
Collapse
Affiliation(s)
- Yanan Hu
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, P. R. China
- Heze Hospital of Traditional Chinese Medicine, Heze, P. R. China
| | - Qiuyang Fan
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, P. R. China
| | - Bo Qiao
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Ou Xu
- Fuwai Yunnan Cardiovascular Hospital, Kunming, P. R. China
| | - Bijun Lv
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, P. R. China
| | - Niping Han
- Molecular Biology for Sinomedicine, Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, P. R. China
| | - Xiaomei Zhang
- Molecular Biology for Sinomedicine, Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, P. R. China
| |
Collapse
|
7
|
Wei J, Tian Y, Wei J, Guan M, Yu X, Xie J, Fan G. Bilirubin regulates cell death type by alleviating macrophage mitochondrial dysfunction caused by cigarette smoke extract. Redox Rep 2024; 29:2382946. [PMID: 39074442 PMCID: PMC11288206 DOI: 10.1080/13510002.2024.2382946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
OBJECTIVES To explore the effects and mechanisms of bilirubin on mitochondrial function and type of macrophage cell death after exposure to cigarette smoke extract (CSE). METHODS RAW264.7 macrophages were treated with different concentrations of CSE and bilirubin solutions and divided into four groups: control, CSE, bilirubin, and bilirubin + CSE groups. The necrotic and apoptotic states of the macrophages were determined using an Annexin V-fluorescein 5-isothiocyanate/propidium iodide (FITC/PI) staining kit. Cytoplasmic NOD-like receptor family, pyrin domain containing 3 (NLRP3) expression in macrophages was detected by immunofluorescence and the levels of IL-1β and IL-18 in the supernatants of culture medium were detected by enzyme linked immunosorbent assay (ELISA) test. A JC-1 mitochondrial membrane potential detection kit was used to assess mitochondrial membrane damage and the adenosine triphosphate (ATP) assay kit was used to determine intracellular ATP levels. After the macrophages were stained with reactive oxygen species (ROS) specific dye, 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA), the fluorescence intensity and proportion of ROS-positive macrophages were measured using flow cytometry. RESULTS We observed that compared with those of 0 μM (control group), concentrations of 5, 10, or 20 μΜ bilirubin significantly decreased cell viability, which was increased by bilirubin exposure below 1 μM. The effect of CSE on macrophage viability was concentration- and time-dependent. Bilirubin of 0.2 μM could alleviate the inhibition of macrophage viability caused by 5% CSE. In addition, bilirubin intervention could reduce the occurrence of necrosis and pyroptosis to a certain extent. CONCLUSIONS CSE could cause mitochondrial dysfunction in macrophages, as demonstrated by a decrease in mitochondrial membrane potential and intracellular ATP levels and an increase in ROS production, while bilirubin could relieve mitochondrial dysfunction caused by CSE.
Collapse
Affiliation(s)
- Jingjing Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Yuan Tian
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jinshu Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Meiqi Guan
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xiaoya Yu
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jianing Xie
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Guoquan Fan
- School of Basic Medical Science, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| |
Collapse
|
8
|
Choi J, Yun SH, Kim H, Lee J, Kim SM, Park MH, Lee HJ, Chun W, Han SB, Ahn KS, Lee JW. Aromadendrin Ameliorates Airway Inflammation in Experimental Mice with Chronic Obstructive Pulmonary Disease. J Microbiol Biotechnol 2024; 35:e2408022. [PMID: 39639498 PMCID: PMC11813338 DOI: 10.4014/jmb.2408.08022] [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: 08/14/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
Aromadendrin (ARO) is an active plant compound that exerts anti-inflammatory effects. However, its ameliorative effects on chronic obstructive pulmonary disease (COPD) remain unclear. Therefore, we investigated the inhibitory effects of ARO on bronchial inflammation using an experimental model of COPD. In vivo analysis confirmed a notable increase in the number of neutrophils/macrophages and the formation of reactive oxygen species (ROS), myeloperoxidase (MPO), interleukin (IL)-6/IL-1β, and monocyte chemoattractant protein (MCP)-1 in the bronchoalveolar lavage (BAL) fluid of COPD mice, which was attenuated by oral gavage of ARO. In addition, hematoxylin and eosin staining showed a notable cell influx in the lungs of the COPD group, which was ameliorated by ARO. Western blotting revealed that ARO decreased the upregulation of neutrophil elastase expression in the lungs of the COPD group. Furthermore, periodic acid-Schiff staining showed that increased mucus formation in the lungs of the COPD group was downregulated by ARO. ARO also blocked CREB activation in the lungs of COPD mice. This in vivo, anti-inflammatory effect of ARO was accompanied by its modulatory effect on the activation of the MAPK/NF-κB/NLRP3 inflammasome. In summary, our study demonstrated that ARO has protective effects on bronchial inflammation by attenuating immune cell accumulation, toxic molecule/cytokine/chemokine formation, and MAPK/NF-κB/NLRP3 inflammasome activation, suggesting the potential development of ARO as an adjuvant for the prevention and treatment of COPD.
Collapse
Affiliation(s)
- Jinseon Choi
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Seok Han Yun
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Hyueyun Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Juhyun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Seong-Man Kim
- O2MEDi Inc. 50 UNIST-gil, Eonyang-eup, Ulju-gun, Ulsan 44919, Republic of Korea
| | - Mi-Hyeong Park
- Office of Surveillance for Narcotics Abuse, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex, Cheongju 28159, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- Department of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
9
|
Zhang P, Gao N, Li X, Zheng X, Kong D, Wu J. Role of m6A Methylation Regulators in the Diagnosis and Subtype Classification of COPD Based on the GEO Database. J Cell Mol Med 2024; 28:e70226. [PMID: 39580709 PMCID: PMC11585962 DOI: 10.1111/jcmm.70226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/30/2024] [Accepted: 11/09/2024] [Indexed: 11/26/2024] Open
Abstract
N6-methyladenosine (m6A) is a prevalent mRNA modifier, yet its role in chronic obstructive pulmonary disease (COPD) remains unexplored. We sourced expression levels of m6A methylation regulators from the GSE76925 dataset. These regulators' differential expression (DEMs) predicted COPD risk via random forest and support vector machine models. Additionally, a nomogram model using DEMs estimated COPD prevalence. We employed consistent cluster analysis of m6A methylation regulators to categorise COPD samples into distinct subtypes. Analyses of immune cell infiltration in these subtypes and differential gene expression (DEGs) across m6A methylation subtypes were conducted. A cell model validated several m6A regulators and their associated pathways. Fifteen m6A methylation regulators showed differential expression and were used in random forest and support vector machine models. Eleven were selected for a nomogram model, which decision curve analysis suggested could benefit patients. Consensus cluster analysis divided the COPD samples into two subtypes: Cluster A and Cluster B. Cluster B was associated with neutrophil and eosinophil-dominated immunity, while Cluster A was linked with monocyte-dominated immunity. Validation of some research findings was achieved through cell experiments. m6A methylation regulators appear instrumental in diagnosing and classifying subtypes of COPD.
Collapse
Affiliation(s)
- Pingan Zhang
- Respiratory Department, the Third Affiliated HospitalBeijing University of Chinese MedicineBeijingChina
| | - Na Gao
- Rehabilitation CollegeZhengzhou Health Vocational CollegeZhengzhouChina
| | - Xiaoning Li
- Respiratory Department, the Third Affiliated HospitalBeijing University of Chinese MedicineBeijingChina
| | - Xudong Zheng
- Respiratory Department, the Third Affiliated HospitalBeijing University of Chinese MedicineBeijingChina
| | - Deyu Kong
- Respiratory Department, the Third Affiliated HospitalBeijing University of Chinese MedicineBeijingChina
| | - Jianjun Wu
- Respiratory Department, the Third Affiliated HospitalBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
10
|
Liang S, Lin J, Xiao M, Shi T, Song Y, Zhang T, Zhou X, Li R, Zhao X, Yang Z, Ti H. Effect of Haoqin Qingdan Tang on influenza A virus through the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155680. [PMID: 38728923 DOI: 10.1016/j.phymed.2024.155680] [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: 11/03/2023] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024]
Abstract
OBJECTIVE Influenza, a viral respiratory illness, leads to seasonal epidemics and occasional pandemics. Given the rising resistance and adverse reactions associated with anti-influenza drugs, Traditional Chinese Medicine (TCM) emerges as a promising approach to counteract the influenza virus. Specifically, Haoqin Qingdan Tang (HQQDT), a TCM formula, has been employed as an adjuvant treatment for influenza in China. However, the active compounds and underlying mechanisms of HQQDT remain unknown. AIM The aim of this study was to investigate HQQDT's antiviral and anti-inflammatory activities in both in vivo and in vitro, and further reveal its active ingredients and mechanism. METHODS In vivo and in vitro experiments were conducted to verify the antiviral and anti-inflammatory activities of HQQDT. Subsequently, the active ingredients and mechanism of HQQDT were explored through combining high performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (HPLC-Q-TOF-MS) analysis and network pharmacology. Finally, the examinations of cell cytokines and signaling pathways aimed to elucidate the predicted mechanisms. RESULTS The results indicated that HQQDT exhibited inhibitory effects on influenza viruses A/PR/8/34 (H1N1), A/HK/1/68 (H3N2), and A/California/4/2009 (H1N1) in vitro. Furthermore, HQQDT enhanced the survival rate of influenza-infected mice, reduced the lung index and lung virus titer, and mitigated lung tissue damage in vivo. The proinflammatory cytokine expression levels upon influenza virus infection in PR8-induced A549 cells or mice were suppressed by HQQDT, including IL-6, IL-1β, CCL2, CCL4, IP-10, interferon β1 (IFN-β1), the interferon regulatory factor 3 (IRF3), and hemagglutinin (HA). Twenty-two active components of HQQDT against influenza were identified using HPLC-Q-TOF-MS analysis. Based on network pharmacological predictions, the JAK/STAT signaling pathway is considered the most relevant for HQQDT's action against influenza. Finally, western blot assays revealed that HQQDT regulated the protein level of the JAK/STAT signaling pathway in PR8-infected A549 cells and lung tissue. CONCLUSION These findings verified the antiviral and anti-inflammatory effects of HQQDT through JAK-STAT signaling pathway in influenza infections, laying the foundation for its further development.
Collapse
Affiliation(s)
- Shiyun Liang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jieling Lin
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Mengjie Xiao
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Institute of Analysis, Guangdong Academy of Sciences(China National Analytical Center, Guangzhou, Guangzhou, 510070, China
| | - Tongmei Shi
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yu Song
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Institute of Analysis, Guangdong Academy of Sciences(China National Analytical Center, Guangzhou, Guangzhou, 510070, China
| | - Tianbo Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xi Zhou
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Institute of Analysis, Guangdong Academy of Sciences(China National Analytical Center, Guangzhou, Guangzhou, 510070, China
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Xin Zhao
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Institute of Analysis, Guangdong Academy of Sciences(China National Analytical Center, Guangzhou, Guangzhou, 510070, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China; Guangzhou Laboratory, Guangzhou, 510000, China; Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou, 510000, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, 519020, China.
| | - Huihui Ti
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Province Precise Medicine Big Date of Traditional Chinese Medicine EngineeringTechnology Research Center, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
11
|
Fan Z, Cui Y, Chen L, Liu P, Duan W. 23-Hydroxybetulinic acid attenuates 5-fluorouracil resistance of colorectal cancer by modulating M2 macrophage polarization via STAT6 signaling. Cancer Immunol Immunother 2024; 73:83. [PMID: 38554148 PMCID: PMC10981607 DOI: 10.1007/s00262-024-03662-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024]
Abstract
Macrophage polarization is closely associated with the inflammatory processes involved in the development and chemoresistance of colorectal cancer (CRC). M2 macrophages, the predominant subtype of tumor-associated macrophages (TAMs) in a wide variety of malignancies, have been demonstrated to promote the resistance of CRC to multiple chemotherapeutic drugs, such as 5-fluorouracil (5-FU). In our study, we investigated the potential of 23-Hydroxybetulinic Acid (23-HBA), a significant active component of Pulsatilla chinensis (P. chinensis), to inhibit the polarization of M2 macrophages induced by IL-4. Our results showed that 23-HBA reduced the expression of M2 specific marker CD206, while downregulating the mRNA levels of M2 related genes (CD206, Arg1, IL-10, and CCL2). Additionally, 23-HBA effectively attenuated the inhibitory effects of the conditioned medium from M2 macrophages on apoptosis in colorectal cancer SW480 cells. Mechanistically, 23-HBA prevented the phosphorylation and nuclear translocation of the STAT6 protein, resulting in the inhibition of IL-10 release in M2 macrophages. Moreover, it interfered with the activation of the IL-10/STAT3/Bcl-2 signaling pathway in SW480 cells, ultimately reducing M2 macrophage-induced resistance to 5-FU. Importantly, depleting STAT6 expression in macrophages abolished the suppressive effect of 23-HBA on M2 macrophage polarization, while also eliminating its ability to decrease M2 macrophage-induced 5-FU resistance in cancer cells. Furthermore, 23-HBA significantly diminished the proportion of M2 macrophages in the tumor tissues of colorectal cancer mice, simultaneously enhancing the anti-cancer efficacy of 5-FU. The findings presented in this study highlight the capacity of 23-HBA to inhibit M2 macrophage polarization, a process that contributes to reduced 5-FU resistance in colorectal cancer.
Collapse
Affiliation(s)
- Zeping Fan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Yaru Cui
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, Jiangxi, China
| | - Lanying Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China.
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China.
- Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, Jiangxi, China.
| | - Peng Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
| | - Wenbin Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, China
- Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
12
|
Li Z, Li X, Feng B, Xue J, Zhao J, Zhu Q, Liu K, Xie F, Xie J. Combining a lung microfluidic chip exposure model with transcriptomic analysis to evaluate the inflammation in BEAS-2B cells exposed to cigarette smoke. Anal Chim Acta 2024; 1287:342049. [PMID: 38182364 DOI: 10.1016/j.aca.2023.342049] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Typically, in vitro studies on the exposure of complex gaseous substances are performed in multi-well plate experiments by trapping and redissolving them, which could introduce potential bias into the results due to the use of inadequate trapping methods. Therefore, a more effective method is to expose complex gaseous substances in gaseous form online, such as using microfluidic chips in experiments. To address these challenges, we introduce a methodology that integrates a self-designed bionic-lung chip with transcriptome analysis to assess the impact of cigarette smoke (CS) exposure on changes in BEAS-2B cells cultured on-chip. RESULTS After the microfluidic chip underwent online gas exposure, total RNA was extracted via in situ cell lysis, and RNA-Seq transcriptome analysis was conducted. And the RNA-Seq findings revealed the significant involvement of the MAPK signaling pathway associated with the inflammatory response in the cellular effects induced by CS exposure. Moreover, the validation of inflammatory response-related biomarkers through in situ fluorescence corroborated the outcomes of the transcriptome analysis. Besides, the experiment involving the inhibition of inflammation by DEX on the microfluidic chip provided additional confirmation of the previous experimental findings. SIGNIFICANT In this study, we present an analytical strategy that combines microfluidic-based CS in situ exposure method with RNA-Seq technology. This strategy offers an experimental scheme for in situ exposure to complex gases, transcriptome analysis, and in situ fluorescence detection. Through the integration of the comprehensiveness of transcriptome analysis with the chip's direct and intuitive in situ fluorescence detection with the stability and reliability of RT-PCR and Western blot experiments, we have successfully addressed the challenges associated with in vitro risk assessment for online exposure to complex gaseous substances.
Collapse
Affiliation(s)
- Zezhi Li
- Beijing Technology and Business University, Beijing 100048, PR China; Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China
| | - Xiang Li
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China; Beijing Life Science Academy, Beijing 102209, PR China.
| | - Boyang Feng
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China
| | - Jingxian Xue
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China
| | - Junwei Zhao
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China; Beijing Life Science Academy, Beijing 102209, PR China
| | - Qingqing Zhu
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China
| | - Kejian Liu
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China
| | - Fuwei Xie
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China
| | - Jianping Xie
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute of CNTC, No. 2 Fengyang Street, Zhengzhou 450001, PR China; Beijing Life Science Academy, Beijing 102209, PR China.
| |
Collapse
|
13
|
He Z, Wang Y, Han L, Hu Y, Cong X. The mechanism and application of traditional Chinese medicine extracts in the treatment of lung cancer and other lung-related diseases. Front Pharmacol 2023; 14:1330518. [PMID: 38125887 PMCID: PMC10731464 DOI: 10.3389/fphar.2023.1330518] [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: 10/31/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Lung cancer stands as one of the most prevalent malignancies worldwide, bearing the highest morbidity and mortality rates among all malignant tumors. The treatment of lung cancer primarily encompasses surgical procedures, radiotherapy, and chemotherapy, which are fraught with significant side effects, unfavorable prognoses, and a heightened risk of metastasis and relapse. Although targeted therapy and immunotherapy have gradually gained prominence in lung cancer treatment, diversifying the array of available methods, the overall recovery and survival rates for lung cancer patients remain suboptimal. Presently, with a holistic approach and a focus on syndrome differentiation and treatment, Traditional Chinese Medicine (TCM) has emerged as a pivotal player in the prognosis of cancer patients. TCM possesses characteristics such as targeting multiple aspects, addressing a wide range of concerns, and minimizing toxic side effects. Research demonstrates that Traditional Chinese Medicine can significantly contribute to the treatment or serve as an adjunct to chemotherapy for lung cancer and other lung-related diseases. This is achieved through mechanisms like inhibiting tumor cell proliferation, inducing tumor cell apoptosis, suppressing tumor angiogenesis, influencing the cellular microenvironment, regulating immune system function, impacting signal transduction pathways, and reversing multidrug resistance in tumor cells. In this article, we offer an overview of the advancements in research concerning Traditional Chinese Medicine extracts for the treatment or adjunctive chemotherapy of lung cancer and other lung-related conditions. Furthermore, we delve into the challenges that Traditional Chinese Medicine extracts face in lung cancer treatment, laying the foundation for the development of diagnostic, prognostic, and therapeutic targets.
Collapse
Affiliation(s)
- Zhenglin He
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Yihan Wang
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Liang Han
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yue Hu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianling Cong
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Gong Q, Yin J, Wang M, Zha C, Yu D, Yang S, Feng Y, Li J, Du L. Anemoside B4 Exerts Hypoglycemic Effect by Regulating the Expression of GLUT4 in HFD/STZ Rats. Molecules 2023; 28:molecules28030968. [PMID: 36770636 PMCID: PMC9921942 DOI: 10.3390/molecules28030968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Anemoside B4 (B4) is a saponin that is extracted from Pulsatilla chinensis (Bge.), and Regel exhibited anti-inflammatory, antioxidant, antiviral, and immunomodulatory activities. However, its hypoglycemic activity in diabetes mellitus has not been evaluated. Here, we explored the effect of B4 on hyperglycemia and studied its underlying mechanism of lowering blood glucose based on hyperglycemic rats in vivo and L6 skeletal muscle cells (L6) in vitro. The rats were fed a high-fat diet (HFD) for one month, combined with an intraperitoneal injection of 60 mg/kg streptozotocin (STZ) to construct the animal model, and the drug was administrated for two weeks. Blood glucose was detected and the proteins and mRNA were expressed. Our study showed that B4 significantly diminished fasting blood glucose (FBG) and improved glucose metabolism. In addition, B4 facilitated glucose utilization in L6 cells. B4 could enhance the expression of glucose transporter 4 (GLUT4) in rat skeletal muscle and L6 cells. Mechanistically, B4 elevated the inhibition of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways. Furthermore, we confirmed the effect of B4 on glucose uptake involved in the enhancement of GLUT4 expression in part due to PI3K/AKT signaling by using a small molecule inhibitor assay and constructing a GLUT4 promoter plasmid. Taken together, our study found that B4 ameliorates hyperglycemia through the PI3K/AKT pathway and promotes GLUT4 initiation, showing a new perspective of B4 as a potential agent against diabetes.
Collapse
Affiliation(s)
- Qin Gong
- School of pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- National Engineering Research Center for Manufacturing Technology of Solid Preparation, Nanchang 330006, China
| | - Jilei Yin
- Institute of Traditional Chinese Medicine, Jiangsu Union Technical Institute Lianyungang Branch, Lianyungang 222007, China
| | - Mulan Wang
- National Engineering Research Center for Manufacturing Technology of Solid Preparation, Nanchang 330006, China
| | - Chengliang Zha
- National Engineering Research Center for Manufacturing Technology of Solid Preparation, Nanchang 330006, China
| | - Dong Yu
- School of pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Shilin Yang
- School of pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- National Engineering Research Center for Manufacturing Technology of Solid Preparation, Nanchang 330006, China
| | - Yulin Feng
- School of pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- National Engineering Research Center for Manufacturing Technology of Solid Preparation, Nanchang 330006, China
| | - Jun Li
- School of pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- National Engineering Research Center for Manufacturing Technology of Solid Preparation, Nanchang 330006, China
- Correspondence: (J.L.); (L.D.); Tel.:+86-180-7009-0101 (J.L.); +791-8711-9785 (L.D.)
| | - Lijun Du
- School of pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, China
- National Engineering Research Center for Manufacturing Technology of Solid Preparation, Nanchang 330006, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Correspondence: (J.L.); (L.D.); Tel.:+86-180-7009-0101 (J.L.); +791-8711-9785 (L.D.)
| |
Collapse
|