1
|
Lee YS, Kwon RJ, Lee HS, Chung JH, Kim YS, Jeong HS, Park SJ, Lee SY, Kim T, Yoon SH. The Role of Pentacyclic Triterpenoids in Non-Small Cell Lung Cancer: The Mechanisms of Action and Therapeutic Potential. Pharmaceutics 2024; 17:22. [PMID: 39861671 PMCID: PMC11768946 DOI: 10.3390/pharmaceutics17010022] [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: 11/30/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Lung cancer remains a major global health problem because of its high cancer-related mortality rate despite advances in therapeutic approaches. Non-small cell lung cancer (NSCLC), a major subtype of lung cancer, is more amenable to surgical intervention in its early stages. However, the prognosis for advanced NSCLC remains poor, owing to limited treatment options. This underscores the growing need for novel therapeutic strategies to complement existing treatments and improve patient outcomes. In recent years, pentacyclic triterpenoids, a group of natural compounds, have emerged as promising candidates for cancer therapy due to their anticancer properties. Pentacyclic triterpenoids, such as lupeol, betulinic acid, betulin, oleanolic acid, ursolic acid, glycyrrhetinic acid, glycyrrhizin, and asiatic acid, have demonstrated the ability to inhibit cell proliferation and angiogenesis, induce apoptosis, suppress metastasis, and modulate inflammatory and immune pathways in NSCLC cell line models. These compounds exert their effects by modulating important signaling pathways such as NF-κB, PI3K/Akt, and MAPK. Furthermore, advances in drug delivery technologies such as nanocarriers and targeted delivery systems have improved the bioavailability and therapeutic efficacy of triterpenoids. However, despite promising preclinical data, rigorous clinical trials are needed to verify their safety and efficacy. This review explores the role of triterpenoids in NSCLC and therapeutic potential in preclinical models, focusing on their molecular mechanisms of action.
Collapse
Affiliation(s)
- Young-Shin Lee
- Family Medicine Clinic and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (Y.-S.L.); (R.J.K.); (H.S.L.)
| | - Ryuk Jun Kwon
- Family Medicine Clinic and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (Y.-S.L.); (R.J.K.); (H.S.L.)
| | - Hye Sun Lee
- Family Medicine Clinic and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (Y.-S.L.); (R.J.K.); (H.S.L.)
| | - Jae Heun Chung
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA;
| | - Yun Seong Kim
- Division of Pulmonology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Han-Sol Jeong
- School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (H.-S.J.); (S.-J.P.); (S.Y.L.)
| | - Su-Jung Park
- School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (H.-S.J.); (S.-J.P.); (S.Y.L.)
| | - Seung Yeon Lee
- School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (H.-S.J.); (S.-J.P.); (S.Y.L.)
| | - Taehwa Kim
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea;
| | - Seong Hoon Yoon
- Division of Pulmonology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| |
Collapse
|
2
|
Chung C, Park SY, Huh JY, Kim NH, Shon C, Oh EY, Park YJ, Lee SJ, Kim HC, Lee SW. Fine particulate matter aggravates smoking induced lung injury via NLRP3/caspase-1 pathway in COPD. J Inflamm (Lond) 2024; 21:13. [PMID: 38654364 DOI: 10.1186/s12950-024-00384-z] [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: 10/22/2022] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Exposure to noxious particles, including cigarette smoke and fine particulate matter (PM2.5), is a risk factor for chronic obstructive pulmonary disease (COPD) and promotes inflammation and cell death in the lungs. We investigated the combined effects of cigarette smoking and PM2.5 exposure in patients with COPD, mice, and human bronchial epithelial cells. METHODS The relationship between PM2.5 exposure and clinical parameters was investigated in patients with COPD based on smoking status. Alveolar destruction, inflammatory cell infiltration, and pro-inflammatory cytokines were monitored in the smoking-exposed emphysema mouse model. To investigate the mechanisms, cell viability and death and pyroptosis-related changes in BEAS-2B cells were assessed following the exposure to cigarette smoke extract (CSE) and PM2.5. RESULTS High levels of ambient PM2.5 were more strongly associated with high Saint George's respiratory questionnaire specific for COPD (SGRQ-C) scores in currently smoking patients with COPD. Combined exposure to cigarette smoke and PM2.5 increased mean linear intercept and TUNEL-positive cells in lung tissue, which was associated with increased inflammatory cell infiltration and inflammatory cytokine release in mice. Exposure to a combination of CSE and PM2.5 reduced cell viability and upregulated NLRP3, caspase-1, IL-1β, and IL-18 transcription in BEAS-2B cells. NLRP3 silencing with siRNA reduced pyroptosis and restored cell viability. CONCLUSIONS PM2.5 aggravates smoking-induced airway inflammation and cell death via pyroptosis. Clinically, PM2.5 deteriorates quality of life and may worsen prognosis in currently smoking patients with COPD.
Collapse
Affiliation(s)
- Chiwook Chung
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Department of Pulmonary and Critical Care Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea
| | - Suk Young Park
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
| | - Jin-Young Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Chung- Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong, Republic of Korea
| | - Na Hyun Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
| | - ChangHo Shon
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Efficacy Evaluation Center, WOOJUNGBIO Inc, Hwaseong, Republic of Korea
| | - Eun Yi Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Jun Park
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seon-Jin Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hwan-Cheol Kim
- Department of Occupational and Environmental Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Xu H, Wen Q, Xu X, Liu Z, Liu S, Wang H, Zhang C, Wan D, Liu K, Du L, Yuan C, Song L. Induction of heme oxygenase-1 antagonizes PM2.5-induced pulmonary VEGFA expression through regulating HIF-1α. J Biochem Mol Toxicol 2023; 37:e23494. [PMID: 37563788 DOI: 10.1002/jbt.23494] [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: 12/15/2022] [Revised: 06/19/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023]
Abstract
Particulate matter (PM) 2.5 has long been regarded as a major risk factor of the respiratory system, which constitutes a threat to human health. Although the positive relationship between PM2.5 exposure and the development of respiratory diseases has been well established, limited studies investigate the intrinsic self-protection mechanisms against PM2.5-induced respiratory injuries. Excessive pulmonary inflammation served as a key pathogenic mechanism in PM2.5-induced airway dysfunction, and we have previously shown that PM2.5 induced the production of vascular endothelial growth factor A (VEGFA) in the bronchial epithelial cells, which subsequently led to pulmonary inflammatory responses. In the current study, we found that PM2.5 also concurrently induced the expression of the stress-responsive protein heme oxygenase-1 (HO-1) along with VEGFA in the bronchial epithelial cells both in vivo and in vitro. Importantly, knocking down of HO-1 expression significantly increased the synthesis and secretion of VEGFA; while overexpression of HO-1 showed the opposite effects, indicating that HO-1 induction can antagonize VEGFA production in the bronchial epithelial cells upon PM2.5 exposure. Mechanistically, HO-1 inhibited PM2.5-evoked VEGFA induction through modulating hypoxia-inducible factor 1 alpha (HIF-1α), which was the upstream transcriptional factor of VEGFA. More specifically, HO-1 could not only inhibit HIF-1α expression, but also suppress its transactivity. Taken together, our results suggested that HO-1 was an intrinsic protective factor against PM2.5-induced pulmonary VEGFA production with a mechanism relating to HIF-1α, thus providing a potential treatment strategy against PM2.5 triggered airway injuries.
Collapse
Affiliation(s)
- Huan Xu
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
- School of Basic Medicine, Anhui Medical University, Hefei, People's Republic of China
| | - Qing Wen
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Xiuduan Xu
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
- School of Basic Medicine, Anhui Medical University, Hefei, People's Republic of China
| | - Zhihui Liu
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
- College of Life Science, Henan Normal University, Xinxiang, People's Republic of China
| | - Shasha Liu
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Hongli Wang
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Henan University, Kaifeng, People's Republic of China
| | - Chongchong Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
- Laboratory of Cellular and Molecular Immunology, School of Medicine, Henan University, Kaifeng, People's Republic of China
| | - Delian Wan
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Kun Liu
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Lina Du
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Chao Yuan
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Lun Song
- Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
- School of Basic Medicine, Anhui Medical University, Hefei, People's Republic of China
- College of Life Science, Henan Normal University, Xinxiang, People's Republic of China
- School of Pharmacy, Jiamusi University, Jiamusi, People's Republic of China
| |
Collapse
|
4
|
Huang D, Jia N, Pei C, Shen Z, Zhao S, Wang Y, Wu Y, Shi S, Li S, Wang Z. Rosavidin protects against PM2.5-induced lung toxicity via inhibition of NLRP3 inflammasome-mediated pyroptosis by activating the PI3K/AKT pathway. Biochem Pharmacol 2023; 213:115623. [PMID: 37244433 DOI: 10.1016/j.bcp.2023.115623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Fine particulate matter (PM2.5) contributes to adverse health effects through the promotion of inflammatory cytokine release. Rosavidin (Ro), a phenylpropanoid compound having multiple biological activities, is extracted from Rhodiola crenulata, a medicine and food homology plant. However, the protective role and mechanism of Ro in PM2.5-induced lung toxicity have not been previously studied. This study aimed to investigate the potential protective effect and mechanism of Ro in PM2.5-induced lung toxicity. A lung toxicity rat model was established through trachea drip of PM2.5 suspension after the different dose pretreatment of Ro (50 mg/kg and 100 mg/kg) to evaluate the effect of Ro on PM2.5 caused lung toxicity. The results showed that Ro attenuated the pathological changes, edema, and inflammation response in rats. The PI3K/AKT signaling pathway may be associated with the protective effect of Ro against pulmonary toxicity. Subsequently, we verified the role of PI3K/AKT in the PM2.5 exposure lung tissue. Moreover, expression levels of p-PI3K and p-AKT were lower, and those of NLRP3, ASC, cleaved caspase-1, cleaved IL-1β, and GSDMD-N were higher in PM2.5 group compared to those in control group. Whereas pre-administration of Ro reversed the expression trends of these proteins in lung tissue. Notably, those protective effects of Ro were not observed after pretreatment with a combination of Ro with nigericin or LY294002. These results indicate that Ro mitigates PM2.5-caused lung toxicity by inhibiting NLRP3 inflammasome-mediated pyroptosis through activation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Demei Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Nan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Caixia Pei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Zherui Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Sijing Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yilan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yongcan Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shihua Shi
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shuiqin Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| | - Zhenxing Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
5
|
Wei X, Lan Y, Nong Z, Li C, Feng Z, Mei X, Zhai Y, Zou M. Ursolic acid represses influenza A virus-triggered inflammation and oxidative stress in A549 cells by modulating the miR-34c-5p/TLR5 axis. Cytokine 2022; 157:155947. [PMID: 35780710 DOI: 10.1016/j.cyto.2022.155947] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ursolic acid (UA) is a pentacyclic triterpenoid compound with a wide range of anti-tumor, anti-inflammatory, hypotensive and other pharmacological effects. Here, the biological roles and regulatory mechanisms of UA in influenza A virus (IAV)-treated A549 cells were investigated. METHOD The cytotoxic impacts of UA on A549 cells with or without IAV treatment were determined using MTT and LDH assays. The inflammatory responses and oxidative stress of IAV-treated A549 cells were measured by RT-qPCR, ELISA, DCFH-DA probe, and colorimetric assays. A dual luciferase assay was carried out to validate the molecular interaction between miR-34c-5p and TLR5. Promoter methylation was detected by MSP experiment. Methylation-related proteins were quantified by western blot. Virus replication was assessed by TCID50 and western blot assays. RESULTS UA significantly ameliorated IAV-triggered cell injury and inflammatory response, virus replication and oxidative stress by elevating cell viability, ROS level and the activities of SOD and GSH-Px but reducing the LDH, MDA, and TCID50 values and the expression of virus-related proteins (NP) and cytokines (TNF-α, IL-1β, IL-6, and IL-18). Moreover, UA promoted miR-34c-5p expression by repressing DNMTs-mediated methylation. TLR5 was verified to be a direct target of miR-34c-5p and could be downregulated by UA. Rescue experiments revealed that silencing miR-34c-5p diminished the regulatory roles of UA in IAV-treated A549 cells. CONCLUSION Our data elucidated that UA attenuated IAV-triggered inflammatory responses and oxidative stress in A549 cells by regulating the miR-34c-5p/TLR5 axis, suggesting that UA plays a protective role in IAV-induced pneumonia.
Collapse
Affiliation(s)
- Xing Wei
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi Province, China
| | - Yuying Lan
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi Province, China
| | - Zhifei Nong
- Department of Pediatrics, Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530022, Guangxi Province, China
| | - Chongjin Li
- Department of Pediatrics, Maoming Hospital of Traditional Chinese Medicine, Maoming 525000, Guangdong Province, China
| | - Zhiqiong Feng
- Department of Pediatrics, Jinshazhou Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Xiaoping Mei
- Department of Pediatrics, Guangxi International Zhuang Medicine Hospital, Nanning 530200, Guangxi Province, China
| | - Yang Zhai
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi Province, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Nanning 530200, Guangxi Province, China; Department of International Medical, Guangxi International Zhuang Medicine Hospital, Nanning 530200, Guangxi Province, China
| | - Min Zou
- Department of Pediatrics, Guangxi International Zhuang Medicine Hospital, Nanning 530200, Guangxi Province, China.
| |
Collapse
|
6
|
Cui X, Zhang Y, Lu Y, Xiang M. ROS and Endoplasmic Reticulum Stress in Pulmonary Disease. Front Pharmacol 2022; 13:879204. [PMID: 35559240 PMCID: PMC9086276 DOI: 10.3389/fphar.2022.879204] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/11/2022] [Indexed: 12/25/2022] Open
Abstract
Pulmonary diseases are main causes of morbidity and mortality worldwide. Current studies show that though specific pulmonary diseases and correlative lung-metabolic deviance own unique pathophysiology and clinical manifestations, they always tend to exhibit common characteristics including reactive oxygen species (ROS) signaling and disruptions of proteostasis bringing about accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER). ER is generated by the unfolded protein response. When the adaptive unfolded protein response (UPR) fails to preserve ER homeostasis, a maladaptive or terminal UPR is engaged, leading to the disruption of ER integrity and to apoptosis, which is called ER stress. The ER stress mainly includes the accumulation of misfolded and unfolded proteins in lumen and the disorder of Ca2+ balance. ROS mediates several critical aspects of the ER stress response. We summarize the latest advances in of the UPR and ER stress in the pathogenesis of pulmonary disease and discuss potential therapeutic strategies aimed at restoring ER proteostasis in pulmonary disease.
Collapse
Affiliation(s)
- Xiangning Cui
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Zhang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yingdong Lu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mi Xiang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Oriola AO, Oyedeji AO. Plant-Derived Natural Products as Lead Agents against Common Respiratory Diseases. Molecules 2022; 27:3054. [PMID: 35630531 PMCID: PMC9144277 DOI: 10.3390/molecules27103054] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/23/2022] [Accepted: 05/07/2022] [Indexed: 12/16/2022] Open
Abstract
Never has the world been more challenged by respiratory diseases (RDs) than it has witnessed in the last few decades. This is evident in the plethora of acute and chronic respiratory conditions, ranging from asthma and chronic obstructive pulmonary disease (COPD) to multidrug-resistant tuberculosis, pneumonia, influenza, and more recently, the novel coronavirus (COVID-19) disease. Unfortunately, the emergence of drug-resistant strains of pathogens, drug toxicity and side effects are drawbacks to effective chemotherapeutic management of RDs; hence, our focus on natural sources because of their unique chemical diversities and novel therapeutic applications. This review provides a summary on some common RDs, their management strategies, and the prospect of plant-derived natural products in the search for new drugs against common respiratory diseases.
Collapse
Affiliation(s)
- Ayodeji Oluwabunmi Oriola
- Department of Chemical and Physical Sciences, Faculty of Natural Sciences, Walter Sisulu University, Nelson Mandela Drive, P/Bag X1, Mthatha 5117, South Africa;
| | | |
Collapse
|
8
|
Luan M, Xu Y, Zhang X, Li D, Yan M, Hou G, Meng Q, Zhao F, Zhao F. Design and synthesis of novel aza-ursolic acid derivatives: in vitro cytotoxicity and nitric oxide release inhibitory activity. Future Med Chem 2022; 14:535-555. [PMID: 35286228 DOI: 10.4155/fmc-2021-0319] [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: 11/17/2022] Open
Abstract
Aim: Inducible nitric oxide synthase (iNOS) is a validated target for anti-inflammatory treatment. Based on the authors' previous work, novel aza-ursolic acid derivatives were designed and synthesized and their inhibitory activities against lipopolysaccharide (LPS)-induced nitric oxide (NO) release from RAW264.7 cells was evaluated. Materials & results: 16 novel derivatives were screened for their in vitro inhibitory activity against NO release using Griess assays and the cytotoxicity was evaluated using MTT assays. The presence of furoxan joined to the A-ring of ursolic acid and N-methylpiperazine groups in the lead compound was identified for anti-inflammatory activity, and compound 21b showed 94.96% inhibition of NO release at 100 μM with an IC50 value of 8.58 μM. Conclusion: Compound 21b has potential anti-inflammatory activity with low cytotoxicity that warrants further preclinical study and evaluation.
Collapse
Affiliation(s)
- Mingzhu Luan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology & Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System & Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Yaoyao Xu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology & Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System & Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Xiaofan Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology & Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System & Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Dalei Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology & Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System & Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Mengjun Yan
- Yantai Raphael Biotechnology Co., Ltd, Yantai, 264043, PR China
| | - Guige Hou
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, PR China
| | - Qingguo Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology & Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System & Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Feng Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology & Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System & Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| | - Fenglan Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology & Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System & Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China
| |
Collapse
|
9
|
Luan M, Wang H, Wang J, Zhang X, Zhao F, Liu Z, Meng Q. Advances in Anti-inflammatory Activity, Mechanism and Therapeutic Application of Ursolic Acid. Mini Rev Med Chem 2022; 22:422-436. [PMID: 34517797 DOI: 10.2174/1389557521666210913113522] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 06/08/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
In vivo and in vitro studies reveal that Ursolic Acid (UA) is able to counteract endogenous and exogenous inflammatory stimuli and has favorable anti-inflammatory effects. The antiinflammatory mechanisms mainly include decreasing the release of histamine in mast cells, suppressing the activities of lipoxygenase, cyclooxygenase and phospholipase, and reducing the production of nitric oxide and reactive oxygen species, blocking the activation of the signal pathway, downregulating the expression of inflammatory factors, and inhibiting the activities of elastase and complement. These mechanisms can open up new avenues for the scientific community to develop or improve novel therapeutic approaches to tackle inflammatory diseases, such as arthritis, atherosclerosis, neuroinflammation, liver diseases, kidney diseases, diabetes, dermatitis, bowel diseases, cancer. The anti-inflammatory activity, the anti-inflammatory mechanism of ursolic acid and its therapeutic applications are reviewed in this paper.
Collapse
Affiliation(s)
- Mingzhu Luan
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, P.R. China
| | - Huiyun Wang
- College of Pharmacy, Jining Medical University, Shandong Province, 276826, P.R. China
| | - Jiazhen Wang
- The Second Hospital of Anhui Medical University, Anhui Province, 230601, P.R. China
| | - Xiaofan Zhang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, P.R. China
| | - Fenglan Zhao
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, P.R. China
| | - Zongliang Liu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, P.R. China
| | - Qingguo Meng
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, P.R. China
| |
Collapse
|
10
|
Romero-Dapueto C, Castillo RL. Oxidative Stress Markers in COPD Patients Admitted to Pulmonary Rehabilitation. Open Respir Med J 2021. [DOI: 10.2174/1874306402115010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background:
Chronic obstructive pulmonary disease (COPD) is a pathology, which leads to an irreversible and progressive reduction of the airflow, usually caused by smoking, but only present in 25% of smokers. Some mechanisms involved in the onset and progression of the disease are local and systemic factors such as inflammation, exacerbated immune response and the appearance of oxidative stress. For all these reasons, the use of oxidative stress parameters as progression markers or even as a way to monitor the response of any kind of non-pharmacological interventions, like the use of pulmonary rehabilitation (PR), is feasible.
Aims:
The study aims to determine markers of oxidative stress levels in plasma and erythrocytes in patients with COPD through the application of a PR protocol.
Methods:
The study included 25 patients diagnosed with COPD according to the GOLD criteria with a medical indication of PR and attendance at the gym in San José Hospital, Santiago, Chile. Blood samples were obtained before the start of the protocol, in the 10th session, and at the end of the protocol (20th session). These samples were stored for oxidative stress determinations: FRAP (ferric reducing ability of plasma), F2-isoprostanes, reduced (GSH)/oxidized (GSSG) ratio and antioxidant enzyme activity in the erythrocyte. In all stages, associations between events and clinical parameters in patients have been observed. The clinical parameters assessed were the six-minute walking test (6MWT), maximal inspiratory and expiratory pressure, the BODE index and Saint George’s respiratory questionnaire, which includes quality of life.
Results:
The intracellular and extracellular capacity (GSH/GSSG and FRAP) in patients in PR at the 10th session were 53.1 and 34% higher than basal values, respectively. Only the GSH/GSSG ratio was 38.2% lower at the 20th session, related in part with higher plasma and erythrocyte lipid peroxidation at baseline. This could be due to the high concentration of reactive oxygen species in the first sessions, which has been reported in the literature as the acute effect of controlled exercise. Blood lipid peroxidation was 43.34 and 58.34% lower at the 10th and 20th sessions, respectively, demonstrating the improvements in the oxidative parameters with long-term exercise. With respect to oxidative enzyme activity, superoxide dismutase and catalase showed higher values of activity at the 10th and 20th sessions compared to the baseline. In the clinical parameters of the PR, significant changes were found in the BODE index and Saint George’s questionnaire, with these results being associated with a less predictive mortality score and a better understanding of the disease. This may be because the patients achieved longer distances in the 6MWT and better understood the disease at the end of the PR.
Conclusion:
The goal of this study was to contribute to the pathophysiological basis for further research on COPD patients, a disease of high prevalence in Chile. This study could support the basis for non-pharmacological strategies such a PR.
Collapse
|
11
|
YG-1 Extract Improves Acute Pulmonary Inflammation by Inducing Bronchodilation and Inhibiting Inflammatory Cytokines. Nutrients 2021; 13:nu13103414. [PMID: 34684415 PMCID: PMC8537401 DOI: 10.3390/nu13103414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 12/23/2022] Open
Abstract
YG-1 extract used in this study is a mixture of Lonicera japonica, Arctic Fructus, and Scutellariae Radix. The present study was designed to investigate the effect of YG-1 extract on bronchodilatation (ex vivo) and acute bronchial and pulmonary inflammation relief (in vivo). Ex vivo: The bronchodilation reaction was confirmed by treatment with YG-1 concentration-accumulation (0.01, 0.03, 0.1, 0.3, and 1 mg/mL) in the bronchial tissue ring pre-contracted by acetylcholine (10 μM). As a result, YG-1 extract is considered to affect bronchodilation by increased cyclic adenosine monophosphate, cAMP) levels through the β2-adrenergic receptor. In vivo: experiments were performed in C57BL/6 mice were divided into the following groups: control group; PM2.5 (fine particulate matter)-exposed group (PM2.5, 200 μg/kg/mL saline); and PM2.5-exposed + YG-1 extract (200 mg/kg/day) group. The PM2.5 (200 μg/kg/mL saline) was exposed for 1 h for 5 days using an ultrasonic nebulizer aerosol chamber to instill fine dust in the bronchi and lungs, thereby inducing acute lung and bronchial inflammation. From two days before PM2.5 exposure, YG-1 extract (200 mg/kg/day) was administered orally for 7 days. The PM2.5 exposure was involved in airway remodeling and inflammation, suggesting that YG-1 treatment improves acute bronchial and pulmonary inflammation by inhibiting the inflammatory cytokines (NLRP3/caspase-1 pathway). The application of YG-1 extract with broncho-dilating effect to acute bronchial and pulmonary inflammation animal models has great significance in developing therapeutic agents for respiratory diseases. Therefore, these results can provide essential data for the development of novel respiratory symptom relievers. Our study provides strong evidence that YG-1 extracts reduce the prevalence of respiratory symptoms and the incidence of non-specific lung diseases and improve bronchial and lung function.
Collapse
|
12
|
Wang J, Wu Q, Ding L, Song S, Li Y, Shi L, Wang T, Zhao D, Wang Z, Li X. Therapeutic Effects and Molecular Mechanisms of Bioactive Compounds Against Respiratory Diseases: Traditional Chinese Medicine Theory and High-Frequency Use. Front Pharmacol 2021; 12:734450. [PMID: 34512360 PMCID: PMC8429615 DOI: 10.3389/fphar.2021.734450] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 12/28/2022] Open
Abstract
Respiratory diseases, especially the pandemic of respiratory infectious diseases and refractory chronic lung diseases, remain a key clinical issue and research hot spot due to their high prevalence rates and poor prognosis. In this review, we aimed to summarize the recent advances in the therapeutic effects and molecular mechanisms of key common bioactive compounds from Chinese herbal medicine. Based on the theories of traditional Chinese medicine related to lung diseases, we searched several electronic databases to determine the high-frequency Chinese medicines in clinical application. The active compounds and metabolites from the selected medicines were identified using the Traditional Chinese Medicine Systems Pharmacology Database (TCMSP) by analyzing oral bioavailability and drug similarity index. Then, the pharmacological effects and molecular mechanisms of the selected bioactive compounds in the viral and bacterial infections, inflammation, acute lung injury (ALI), chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, asthma, and lung cancer were summarized. We found that 31 bioactive compounds from the selected 10 common Chinese herbs, such as epigallocatechin-3-gallate (EGCG), kaempferol, isorhamnetin, quercetin, and β-sitosterol, can mainly regulate NF-κB, Nrf2/HO-1, NLRP3, TGF-β/Smad, MAPK, and PI3K/Akt/mTOR pathways to inhibit infection, inflammation, extracellular matrix deposition, and tumor growth in a series of lung-related diseases. This review provides novel perspectives on the preclinical study and clinical application of Chinese herbal medicines and their bioactive compounds against respiratory diseases.
Collapse
Affiliation(s)
- Jing Wang
- Department of Respiratory, Changchun University of Chinese Medicine, Changchun, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lu Ding
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Siyu Song
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yaxin Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Li Shi
- Department of Respiratory, Changchun University of Chinese Medicine, Changchun, China
| | - Tan Wang
- Department of Respiratory, Changchun University of Chinese Medicine, Changchun, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zeyu Wang
- Department of Scientific Research, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Jilin Ginseng Academy, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
13
|
Jia H, Liu Y, Guo D, He W, Zhao L, Xia S. PM2.5-induced pulmonary inflammation via activating of the NLRP3/caspase-1 signaling pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:298-307. [PMID: 32996690 PMCID: PMC7891361 DOI: 10.1002/tox.23035] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 05/07/2023]
Abstract
Particulate matter 2.5 (PM2.5)-induced pulmonary inflammation has become a public concern in recent years. In which, the activation of the NLRP3/caspase-1 pathway was closely related to the inflammatory response of various diseases. However, the promotion effect of the NLRP3/caspase-1 pathway on PM2.5-induced pulmonary inflammation remains largely unclear. Here, our data showed that PM2.5 exposure caused lung injury in the mice by which inflammatory cell infiltration occurred in lung and alveolar structure disorder. Meanwhile, the exposure of human bronchial epithelial cells (16HBE) to PM2.5 resulted in suppressed cell viability, as well as elevated cell apoptosis. Moreover, a higher level of inflammatory cytokine and activation of the NLRP3/caspase-1 pathway in PM2.5-induced inflammation mice models and 16HBE cells. Mechanistically, pretreatment with MCC950, a NLRP3/caspase-1 pathway inhibitor, prevented PM2.5-induced lung injury, inflammatory response, and the number of inflammatory cells in BALFs, as well as promoted cell viability and decreased inflammatory cytokine secretion. Collectively, our findings indicated that the NLRP3/caspase-1 pathway serves a vital role in the pathological changes of pulmonary inflammation caused by PM2.5 exposure. MCC950 was expected to be the therapeutic target of PM2.5 inhalation mediated inflammatory diseases.
Collapse
Affiliation(s)
- Hui Jia
- Department of Respiratory and Critical Care MedicineCentral Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Yang Liu
- Department of Respiratory and Critical Care MedicineCentral Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Dan Guo
- Department of Respiratory and Critical Care MedicineCentral Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Wei He
- Department of Respiratory and Critical Care MedicineCentral Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Long Zhao
- Department of Respiratory and Critical Care MedicineCentral Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Shuyue Xia
- Department of Respiratory and Critical Care MedicineCentral Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| |
Collapse
|
14
|
Yue Q, Deng X, Li Y, Zhang Y. Effects of Betulinic Acid Derivative on Lung Inflammation in a Mouse Model of Chronic Obstructive Pulmonary Disease Induced by Particulate Matter 2.5. Med Sci Monit 2021; 27:e928954. [PMID: 33612710 PMCID: PMC7885291 DOI: 10.12659/msm.928954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is mainly induced by the increased content of particulate matter 2.5 (PM2.5) in the atmosphere. This study aimed to evaluate the effects of betulinic acid derivative on lung inflammation in a mouse model of chronic obstructive pulmonary disease induced by particulate matter 2.5. MATERIAL AND METHODS The mice were given a PM2.5 (25 μl) suspension for 7 days by the intranasal route to establish a COPD model. The content of TNF-alpha and IL-6 in the BALF samples was measured by commercially available ELISA kits. RESULTS The PM2.5-induced higher LDH and ACP levels were significantly alleviated in mouse lung tissues by treatment with betulinic acid derivative. Treatment with betulinic acid derivative also suppressed PM2.5-induced increase in AKP and ALB levels in mouse lung tissues. Betulinic acid derivative reversed PM2.5-mediated suppression of SOD activity and elevation of NOS level in mouse BALF. Moreover, the PM2.5-induced excessive NO and MDA levels in mouse BALF were significantly reduced (P.
Collapse
Affiliation(s)
- Qianyu Yue
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Xiaoli Deng
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Yuntao Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Yunhui Zhang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| |
Collapse
|
15
|
Ren C, Kong D, Ning C, Xing H, Cheng Y, Zhang Y, Lu Y, Li N, Chen X, Zhao D. Improved Pharmacokinetic Characteristics of Ursolic Acid in Rats Following Intratracheal Instillation and Nose-Only Inhalation Exposure. J Pharm Sci 2021; 110:905-913. [PMID: 33049258 DOI: 10.1016/j.xphs.2020.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/16/2022]
Abstract
Ursolic acid (UA) is a common pentacyclic triterpene phytochemical with various pharmacological activities. However, UA is classified as a class IV drug in BCS system and its development as an oral drug is limited. Pulmonary delivery is an effective way to improve the bioavailability of drugs with low absorption. In this study, the differences in pharmacokinetic behaviors of UA after pulmonary and oral administration was explored in rats. Compared with oral administration, the plasma concentration of UA increased rapidly after pulmonary administration, and the bioavailability increased about 80 times. UA instantly accumulated in the lungs after pulmonary administration, and the pulmonary AUC0-t/dose increased by 114 times compared to oral dosing. Incubation experiments showed that the metabolism of UA in rat lung microsomes was significantly reduced compared with that in liver microsomes, in which the clearance rate of phase I and phase II metabolism was reduced by 14.7 times and 1.4 times respectively. These results indicated that pulmonary administration could improve the bioavailability of UA and reduce its metabolism. This study not only provides a preferable route of administration for the application of UA but also offers new insights for the development of phytochemical drug candidates with poor pharmacokinetic properties.
Collapse
Affiliation(s)
- Chang Ren
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Dexuan Kong
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chen Ning
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Han Xing
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yujie Cheng
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yongjie Zhang
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Lu
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ning Li
- National Experimental Teaching Demonstration Center of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Di Zhao
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
16
|
Gao J, Wei Q, Pan R, Yi W, Xu Z, Duan J, Tang C, He Y, Liu X, Song S, Su H. Elevated environmental PM 2.5 increases risk of schizophrenia relapse: Mediation of inflammatory cytokines. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 753:142008. [PMID: 32892002 DOI: 10.1016/j.scitotenv.2020.142008] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/13/2020] [Accepted: 08/25/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Ecological epidemiology suggests that hospital admissions for schizophrenia are associated with an increased environmental PM2.5, but no prospective study has verified this result, and the physiological mechanism is not clear. METHODS We used a repeated-measures design to prospectively assess the association of environmental PM2.5 and the risk of relapse in schizophrenia, and used two linear mixed-effects models to explore possible mediating effects of immune cytokines on the premise of controlling confounders. RESULTS We import the data using EpiData software, and collate and analyze of the data using R software. The increase of PM2.5 at lag0 had the greatest impact on the relapse of schizophrenia (for each 10 μg/m3 increase in PM2.5, the relapse risk score increased by 1.504, that is to say, odds ratio (OR) = 4.500 (95% confidence interval (CI): 2.849-7.106,P < 0.001)), and cumulative effects lasted for four days with the maximum at the second day (for each 10 μg/m3 increase in PM2.5, the relapse risk score increased by 1.301, OR = 3.673 (95%CI: 1.962-6.876,P < 0.001)). PM2.5 exposure was statistically related to four symptom dimensions of early signs scale (ESS), and the symptoms most affected by the increased PM2.5 were depression/withdrawal (ESSN) (OR = 1.990, 95%CI: 1.701-2.328), anxiety/agitation (ESS-A) (OR = 1.537, 95%CI: 1.340-1.763), initial psychosis (ESS-IP) (OR = 1.398, 95%CI: 1.151-1.697), and disinhibition (ESS-D) (OR = 1.235, 95%CI: 1.133-1.347). Furthermore, there are three statistically significant pathways in intermediary analysis: of PM2.5 and relapse risk: "PM2.5 → IL-17 → ESS", "PM2.5 → IL-17 → ESS-A", and "PM2.5 → IL-17 → ESS-N", and the intermediary ratio of IL-17 was 11.66%, 16.37% and 22.55%, respectively. CONCLUSIONS Increased environmental PM2.5 is a risk factor for the relapse of schizophrenia. Early relapse identification and intervention based on clinical characteristics are of great significance for timely termination of relapse and slowing down of relapse.
Collapse
Affiliation(s)
- Jiaojiao Gao
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Qiannan Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Rubing Pan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Weizhuo Yi
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Zihan Xu
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Jun Duan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Chao Tang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Yangyang He
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Xiangguo Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Shasha Song
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China
| | - Hong Su
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Major Autoimmune Disease, China.
| |
Collapse
|
17
|
Liu G, Li J, Shi L, Liu M, Cai B. Advances in the Study of Structural Modification and Biological Activities of Ursolic Acid. CHINESE J ORG CHEM 2021. [DOI: 10.6023/cjoc202102032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
18
|
Wang M, Theis T, Kabat M, Loers G, Agre LA, Schachner M. Functions of Small Organic Compounds that Mimic the HNK-1 Glycan. Int J Mol Sci 2020; 21:ijms21197018. [PMID: 32987628 PMCID: PMC7582369 DOI: 10.3390/ijms21197018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 12/27/2022] Open
Abstract
Because of the importance of the HNK-1 carbohydrate for preferential motor reinnervation after injury of the femoral nerve in mammals, we screened NIH Clinical Collection 1 and 2 Libraries and a Natural Product library comprising small organic compounds for identification of pharmacologically useful reagents. The reason for this attempt was to obviate the difficult chemical synthesis of the HNK-1 carbohydrate and its isolation from natural sources, with the hope to render such compounds clinically useful. We identified six compounds that enhanced neurite outgrowth from cultured spinal motor neurons at nM concentrations and increased their neurite diameter, but not their neurite branch points. Axons of dorsal root ganglion neurons did not respond to these compounds, a feature that is in agreement with their biological role after injury. We refer to the positive functions of some of these compounds in animal models of injury and delineate the intracellular signaling responses elicited by application of compounds to cultured murine central nervous system neurons. Altogether, these results point to the potential of the HNK-1 carbohydrate mimetics in clinically-oriented settings.
Collapse
Affiliation(s)
- Minjuan Wang
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.W.); (T.T.); (M.K.)
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.W.); (T.T.); (M.K.)
| | - Maciej Kabat
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.W.); (T.T.); (M.K.)
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Lynn A. Agre
- Rutgers School of Arts and Sciences, Department of Statistics and Rutgers Business School, Rutgers University, Piscataway, NJ 08854, USA;
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.W.); (T.T.); (M.K.)
- Correspondence: ; Tel.: +1-848-445-1780
| |
Collapse
|
19
|
Mao M, Li J, Bi A, Jia H, Li Q, Liu Y, Jiang X, Huang D, Xia S. Thymoquinone ameliorates the PM2.5-induced lung injury in rats. Exp Lung Res 2020; 46:297-307. [PMID: 32748670 DOI: 10.1080/01902148.2020.1801895] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND This study aims to explore the effect of thymoquinone (TQ) on particulate matter 2.5 (PM2.5)-induced lung injury. METHODS The PM2.5 sample was provided by Shenyang Environment Monitor Central Station. Lung injury was established by intratracheal instillation PM2.5 (7.5 mg/kg) followed by TQ treatment (20 and 40 mg/kg) for 14 d in rats. Hematoxylin and eosin (HE) and Evans blue dye (EBD) staining were detected on lung tissues. ELISA, real-time PCR, western blotting and TUNEL assays were also performed. RESULTS The data showed that TQ diminished lung injury and EBD accumulation. The number of macrophages, neutrophils, eosinophils, and lymphocytes was ameliorated after TQ treatment. In addition, TQ suppressed the inflammation reaction parameters (interleukin-1β and -6, IL-1β and IL-6; tumor necrosis factor-α, TNF-α) and oxidative stress in PM2.5-induced lung injury. The levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase (HO-1) were increased due to the treatment of TQ. The number of TUNEL-positive cells was prominently reduced in TQ-treated rats compared with that in PM2.5 group. Intratracheal instillation PM2.5 activated autophagy, whilst TQ blocked it in lung. CONCLUSIONS Taken together, this study provides the first in vivo evidence that TQ suppresses inflammation, oxidative stress, apoptosis, and autophagy in PM2.5-induced lung injury.
Collapse
Affiliation(s)
- Mingqing Mao
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, People's Republic of China
| | - Jing Li
- Shenyang Environment Monitor Central Station, Key Laboratory of Atmospheric Organic Compound Monitoring and Analysis, Ministry of Environmental Protection, Shenyang, People's Republic of China
| | - Aiping Bi
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, People's Republic of China
| | - Hui Jia
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, People's Republic of China
| | - Qiong Li
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, People's Republic of China
| | - Yang Liu
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, People's Republic of China
| | - Xiaochuan Jiang
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, People's Republic of China
| | - Desheng Huang
- Department of Mathematics, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Shuyue Xia
- Department of Respiratory and Critical Care Medicine, Central Hospital Affiliated to Shenyang Medical College, Shenyang, People's Republic of China
| |
Collapse
|
20
|
Shawky E, Nada AA, Ibrahim RS. Potential role of medicinal plants and their constituents in the mitigation of SARS-CoV-2: identifying related therapeutic targets using network pharmacology and molecular docking analyses. RSC Adv 2020; 10:27961-27983. [PMID: 35519104 PMCID: PMC9055652 DOI: 10.1039/d0ra05126h] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/17/2020] [Indexed: 12/18/2022] Open
Abstract
Since the outbreak of Coronavirus disease (COVID-19) caused by SARS-CoV-2 in December 2019, there has been no vaccine or specific antiviral medication for treatment of the infection where supportive care and prevention of complications is the current management strategy. In this work, the potential use of medicinal plants and more than 16 500 of their constituents was investigated within two suggested therapeutic strategies in the fight against SARS-CoV-2 including prevention of SARS-CoV-2 RNA synthesis and replication, through targeting vital proteins and enzymes as well as modulation of the host's immunity through production of virulence factors. Molecular docking studies on the viral enzymes 3Clpro, PLpro and RdRp suggested rocymosin B, verbascoside, rutin, caftaric acid, luteolin 7-rutinoside, fenugreekine and cyanidin 3-(6′′-malonylglucoside) as promising molecules for further drug development. Meanwhile, the medicinal plants Glycyrrhiza glabra, Hibiscus sabdariffa, Cichorium intybus, Chrysanthemum coronarium, Nigella sativa, Anastatica hierochuntica, Euphorbia species, Psidium guajava and Epilobium hirsutum were enriched in compounds with the multi-targets PTGS2, IL2, IL1b, VCAM1 and TNF such as quercetin, ursolic acid, kaempferol, isorhamnetin, luteolin, glycerrhizin and apigenin. Enriched pathways of the molecular targets included cytokine–cytokine receptor interaction, TNF signaling pathway, NOD-like receptor signaling pathway, Toll-like receptor signaling pathway, NF-kappa B signaling pathway and JAK-STAT3 signaling pathway which are all closely related to inflammatory, innate and adaptive immune responses. The present study identified natural compounds targeting SARS-CoV-2 for further in vitro and in vivo studies and emphasizes the potential role of medicinal plants in the mitigation of SARS-CoV-2. Since the outbreak of Coronavirus disease (COVID-19) caused by SARS-CoV-2 in December 2019, there has been no vaccine or specific antiviral medication for treatment of the infection where supportive care and prevention of complications is the current management strategy.![]()
Collapse
Affiliation(s)
- Eman Shawky
- Department of Pharmacognosy
- Faculty of Pharmacy
- Alexandria University
- Alexandria 21521
- Egypt
| | - Ahmed A. Nada
- Department of Pharmacognosy
- Faculty of Pharmacy
- Alexandria University
- Alexandria 21521
- Egypt
| | - Reham S. Ibrahim
- Department of Pharmacognosy
- Faculty of Pharmacy
- Alexandria University
- Alexandria 21521
- Egypt
| |
Collapse
|