1
|
Wang L, Chen H, Deng L, Hu M, Wang Z, Zhang K, Lian C, Wang X, Zhang J. Roburic acid inhibits lung cancer metastasis and triggers autophagy as verified by network pharmacology, molecular docking techniques and experiments. Front Oncol 2024; 14:1449143. [PMID: 39450260 PMCID: PMC11499198 DOI: 10.3389/fonc.2024.1449143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Background Roburic acid (ROB) is a newly discovered tetracyclic triterpene acid extracted from oak galls, which has anti-inflammatory effects, but the mechanism of its anticancer effect is not clear. Our study focuses on exploring the potential mechanism of action of ROB in the treatment of lung cancer using a combination of network pharmacological prediction, molecular docking technique and experimental validation. Methods A network pharmacology approach was used to screen the protein targets of ROB and lung cancer, and PPI network analysis and enrichment analysis were performed on the intersecting genes. The tissue and organ distribution of the targets was also evaluated based on the BioGPS database. To ensure the reliability of the network pharmacology prediction results, we proceeded to use molecular docking technique to determine the relationship between drugs and targets. Finally, in vitro experiments with cell lines were performed to further reveal the potential mechanism of ROB for the treatment of lung cancer. Results A total of 83 potential targets of ROB in lung cancer were collected and further screened by using Cytoscape software, and 7 targets of PTGS2, CYP19A1, PTGS1, AR, CYP17A1, PTGES and SRD5A1 were obtained as hub genes and 7 hub targets had good binding energy with ROB. GO and KEGG analysis showed that ROB treatment of lung cancer mainly involves Arachidonic acid metabolism, Notch signaling pathway, cancer pathway and PPAR signaling pathway. The results of in vitro experiments indicated that ROB may inhibit the proliferation and metastasis of lung cancer cells and activate the PPARγ signaling pathway, as well as induce cellular autophagy. Conclusions The results of this study comprehensively elucidated the potential targets and molecular mechanisms of ROB for the treatment of lung cancer, providing new ideas for further lung cancer therapy.
Collapse
Affiliation(s)
- Luyao Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Lili Deng
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Mengling Hu
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Ziqiang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Kai Zhang
- Research Center of Clinical Medicine, Bengbu Medical University, Bengbu, China
| | - Chaoqun Lian
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Joint Research Center for Regional Diseases of Institute of Healthcare Management (IHM), The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, China
| |
Collapse
|
2
|
Wang L, Wang Z, Ni Y, Wang X, Zhang T, Hu M, Lian C, Wang X, Zhang J. Elucidating the mechanism of action of Isobavachalcone induced autophagy and apoptosis in non-small cell lung cancer by network pharmacology and experimental validation methods. Gene 2024; 918:148474. [PMID: 38670393 DOI: 10.1016/j.gene.2024.148474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer deaths, and non-small cell lung cancer (NSCLC) accounts for the majority of lung cancer-related mortality. In recent years, there have been numerous treatments for non-small cell lung cancer, but the cure and survival rates are still extremely low. Isobavachalcone (IBC) belongs to the chalcone component of the traditional Chinese medicine Psoralea corylifolia L., and is a unique Protein kinase B (AKT) pathway inhibitor with significant anticancer effects. Previous studies have shown that IBC possess a variety of biological properties, including anti-cancer, anti-inflammatory, and antioxidant properties. This study focused on the use of network pharmacology analysis, molecular docking technology and experimental validation to elucidate the potential mechanisms of IBC for the treatment of NSCLC. METHODS Screening key genes and pathways of IBC action in NSCLC using network pharmacology. The IBC target genes were from The Encyclopedia of Traditional Chinese Medicine (ETCM) and BATMAN-TCM databases, the NSCLC target genes were from GeneCards, Online Mendelian Inheritance in Man (OMIM) and The Therapeutic Target database (TTD) databases, both of which were taken as intersecting genes for protein-protein interaction network analysis and enrichment analysis, and the binding energies of the compounds to the core targets were further verified by molecular docking. Cell lines in vitro experiments were then performed to further unravel the mechanism of IBC for NSCLC. RESULTS A total of 279 potential targets were retrieved by searching the intersection of IBC and NSCLC targets. Protein-protein interaction (PPI) network analysis indicated that 6 targets, including AKT1, RXRA, NCOA1, RXRB, RARA, PPARG were hub genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis suggested that IBC treatment of NSCLC mainly involves steroid binding, transcription factor activity, Pathways in cancer, cAMP signaling pathway, Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway. Among them, the AMPK signaling pathway, which contained the largest number of enriched genes, may play a greater role in the treatment of NSCLC. Then, the results of in vitro experiment indicated that IBC could inhibit proliferation of NSCLC cells and induce cell autophagy and apoptosis. The results also showed that IBC could increase the protein expression of AMPK and decrease the protein expression of AKT and mammalian target of rapamycin (mTOR), suggesting that IBC can treat NSCLC by inducing cellular autophagy and apoptosis as well as modulating AMPK and AKT signaling pathways. CONCLUSIONS In summary, this study provided a new insight into the protective mechanism of IBC against NSCLC through network pharmacology and experimental validation.
Collapse
Affiliation(s)
- Luyao Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233030, China; Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Ziqiang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu 233030, China
| | - Yuhan Ni
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233030, China
| | - Xue Wang
- Digestive Department, Xi'an Fifth Hospital, Xi'an 710000, China
| | - Tingting Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Mengling Hu
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233030, China
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu 233030, China.
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233030, China; Joint Research Center for Regional Diseases of IHM, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China.
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233030, China.
| |
Collapse
|
3
|
Liu R, Jiang L, Chen Y, Shao J, Chen K, Li X, Lv J, Cai W, Cai H, Zhu Z, Wang C, Zhou K, Huang J, Xiao J, Ni W, Wu C. Ginsenoside-Rh2 Promotes Functional Recovery after Spinal Cord Injury by Enhancing TFEB-Mediated Autophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14727-14746. [PMID: 38907713 DOI: 10.1021/acs.jafc.4c02379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Background: Following spinal cord injury (SCI), autophagy plays a positive role in neuronal protection, whereas pyroptosis triggers an inflammatory response. Ginsenoside-Rh2 (GRh2), known for its neuroprotective effects, is considered a promising drug. However, the exact molecular mechanisms underlying these protective effects remain unclear. Aim of the Study: Explore the therapeutic value of GRh2 in SCI and its potential mechanisms of action. Materials and Methods: An SCI mouse model was established, followed by random grouping and drug treatments under different conditions. Subsequently, the functional recovery of SCI mice after GRh2 treatment was assessed using hematoxylin and eosin, Masson's trichrome, and Nissl staining, footprint analysis, Basso Mouse Scale scoring, and inclined plane tests. The expression levels of relevant indicators in the mice were detected using Western blotting, immunofluorescence, and a quantitative polymerase chain reaction. Network pharmacology analysis was used to identify the relevant signaling pathways through which GRh2 exerts its therapeutic effects. Results: GRh2 promoted functional recovery after SCI. GRh2 significantly inhibits pyroptosis by enhancing autophagy in SCI mice. Simultaneously, the neuroprotective effect of GRh2, achieved through the inhibition of pyroptosis, is partially reversed by 3-methyladenine, an autophagy inhibitor. Additionally, the increase in autophagy induced by GRh2 is mediated by the promotion of transcription factor EB (TFEB) nuclear translocation and dephosphorylation. Partial attenuation of the protective effects of GRh2 was observed after TFEB knockdown. Additionally, GRh2 can modulate the activity of TFEB in mice post-SCI through the EGFR-MAPK signaling pathway, and NSC228155 (an EGFR activator) can partially reverse the effect of GRh2 on the EGFR-MAPK signaling pathway. Conclusions: GRh2 improves functional recovery after SCI by upregulating TFEB-mediated autophagic flux and inhibiting pyroptosis, indicating its potential clinical applicability.
Collapse
Affiliation(s)
- Rongjie Liu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Liting Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Yituo Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Jiaqin Shao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Kongbin Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiang Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Junlei Lv
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Wanta Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxu Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhefan Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenggui Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Jinfeng Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wenfei Ni
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenyu Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
4
|
Dana SMMA, Meghdadi M, Kakhki SK, Khademi R. Anti-leukemia effects of ginsenoside monomer: A narrative review of pharmacodynamics study. CURRENT THERAPEUTIC RESEARCH 2024; 100:100739. [PMID: 38706463 PMCID: PMC11066596 DOI: 10.1016/j.curtheres.2024.100739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/12/2024] [Indexed: 05/07/2024]
Abstract
Background Leukemia is a prevalent disease with high mortality and morbidity rates. Current therapeutic approaches are expensive and have side effects. Objective In this investigation, we reviewed studies that investigated the anticancer effects of ginsenoside derivatives against leukemia and also explained the three main Ginsenoside derivatives (ginsenoside Rg3, Rh2, and Rg1) separately. Methods An extensive search was conducted in Pubmed, Web of Science, and Google Scholar and relevant studies that investigated anticancer effects of ginsenoside derivatives against leukemia cancer were extracted and reviewed. Results Preclinical studies reported that ginsenoside derivatives can induce apoptosis, suppress the proliferation of cancer cells, and induce differentiation and cell cycle arrest in leukemia cells. in addition, it can suppress the chemokine activity and extramedullary infiltration of leukemia cells from bone marrow. using herbal medicine and its derivatives is a promising approach to current health problems. Conclusion This review shows that ginsenoside derivatives can potentially suppress the growth of leukemia cells via various pathways and can be applied as a new natural medicine for future clinical research.
Collapse
Affiliation(s)
| | - Mohammadreza Meghdadi
- Department of Hematology and Blood Banking, Faculty of Medical Science, Mashhad University of Medical Science, Mashhad, Iran
| | - Saeed Khayat Kakhki
- Department of Gerontological Nursing, School of Nursing, Social Development and Health Promotion Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Reza Khademi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Jung YY, Ahn KS, Shen M. Unveiling autophagy complexity in leukemia: The molecular landscape and possible interactions with apoptosis and ferroptosis. Cancer Lett 2024; 582:216518. [PMID: 38043785 DOI: 10.1016/j.canlet.2023.216518] [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: 11/03/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
Autophagy is a self-digestion multistep process in which causes the homeostasis through degradation of macromolecules and damaged organelles. The autophagy-mediated tumor progression regulation has been a critical point in recent years, revealing the function of this process in reduction or acceleration of carcinogenesis. Leukemia is a haematological malignancy in which abnormal expansion of hematopoietic cells occurs. The current and conventional therapies from chemotherapy to cell transplantation have failed to appropriately treat the leukemia patients. Among the mechanisms dysregulated in leukemia, autophagy is a prominent one in which can regulate the hallmarks of this tumor. The protective autophagy inhibits apoptosis and ferroptosis in leukemia, while toxic autophagy accelerates cell death. The proliferation and invasion of tumor cells are tightly regulated by the autophagy. The direction of regulation depends on the function of autophagy that is protective or lethal. The protective autophagy accelerates chemoresistance and radio-resistsance. The non-coding RNAs, histone transferases and other pathways such as PI3K/Akt/mTOR are among the regulators of autophagy in leukemia progression. The pharmacological intervention for the inhibition or induction of autophagy by the compounds including sesamine, tanshinone IIA and other synthetic compounds can chance progression of leukemia.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Mingzhi Shen
- Department of General Medicine, Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Sanya, China.
| |
Collapse
|
6
|
Thapa R, Gupta G, Bhat AA, Almalki WH, Alzarea SI, Kazmi I, Saleem S, Khan R, Altwaijry N, Dureja H, Singh SK, Dua K. A review of Glycogen Synthase Kinase-3 (GSK3) inhibitors for cancers therapies. Int J Biol Macromol 2023; 253:127375. [PMID: 37839597 DOI: 10.1016/j.ijbiomac.2023.127375] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
The intricate molecular pathways governing cancer development and progression have spurred intensive investigations into novel therapeutic targets. Glycogen Synthase Kinase-3 (GSK3), a complex serine/threonine kinase, has emerged as a key player with intricate roles in various cellular processes, including cell proliferation, differentiation, apoptosis, and metabolism. Harnessing GSK3 inhibitors as potential candidates for cancer therapy has garnered significant interest due to their ability to modulate key signalling pathways that drive oncogenesis. The review encompasses a thorough examination of the molecular mechanisms underlying GSK3's involvement in cancer progression, shedding light on its interaction with critical pathways such as Wnt/β-catenin, PI3K/AKT, and NF-κB. Through these interactions, GSK3 exerts influence over tumour growth, invasion, angiogenesis, and metastasis, rendering it an attractive target for therapeutic intervention. The discussion includes preclinical and clinical studies, showcasing the inhibitors efficacy across a spectrum of cancer types, including pancreatic, ovarian, lung, and other malignancies. Insights from recent studies highlight the potential synergistic effects of combining GSK3 inhibitors with conventional chemotherapeutic agents or targeted therapies, opening avenues for innovative combinatorial approaches. This review provides a comprehensive overview of the current state of research surrounding GSK3 inhibitors as promising agents for cancer treatment.
Collapse
Affiliation(s)
- Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India.
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shakir Saleem
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Ruqaiyah Khan
- Department of Basic Health Sciences, Deanship of Preparatory Year for the Health Colleges, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Najla Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Harish Dureja
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
7
|
Zhang R, Xu S, Yuan M, Guo L, Xie L, Liao Y, Xu Y, Fu X. An ultrasmall PVP-Fe-Cu-Ni-S nano-agent for synergistic cancer therapy through triggering ferroptosis and autophagy. NANOSCALE 2023; 15:12598-12611. [PMID: 37462439 DOI: 10.1039/d3nr02708b] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Photothermal therapy (PTT) is an emerging field where photothermal agents could convert visible or near-infrared (NIR) radiation into heat to kill tumor cells. However, the low photothermal conversion efficiency of photothermal agents and their limited antitumor activities hinder the development of these agents into monotherapies for cancer. Herein, we have fabricated an ultrasmall polyvinylpyrrolidone (PVP)-Fe-Cu-Ni-S (PVP-NP) nano-agent via a simple hot injection method with excellent photothermal conversion efficiency (∼96%). Photothermal therapy with this nano-agent effectively inhibits tumor growth without apparent toxic side-effects. Mechanistically, our results demonstrated that, after NIR irradiation, PVP-NPs can induce ROS/singlet oxygen generation, decrease the mitochondrial membrane potential, release extracellular Fe2+, and consume glutathione, triggering autophagy and ferroptosis of cancer cells. Moreover, PVP-NPs exhibit excellent contrast enhancement according to magnetic resonance imaging (MRI) analysis. In summary, PVP-NPs have a high photothermal conversion efficiency and can be applied for MRI-guided synergistic photothermal/photodynamic/chemodynamic cancer therapy, resolving the bottleneck of existing phototherapeutic agents.
Collapse
Affiliation(s)
- Rongjun Zhang
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Shuxiang Xu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China.
- Binjiang Research Institute of Zhejiang University, Hangzhou, Zhejiang 310052, China
| | - Miaomiao Yuan
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Lihao Guo
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Luoyijun Xie
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Yingying Liao
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Yang Xu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China.
- Binjiang Research Institute of Zhejiang University, Hangzhou, Zhejiang 310052, China
| | - Xuemei Fu
- International Peace Maternity and Child Health Hospital of China Welfare Institution, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
8
|
Qi X, Sun X, Wang M, Wang M, Qi Z, Cui C. Ginseng polysaccharides ameliorate abnormal lipid metabolism caused by acute alcoholic liver injury by promoting autophagy. FOOD FRONTIERS 2023. [DOI: 10.1002/fft2.204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Xin Qi
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education Yanbian University Yanji China
| | - Xihan Sun
- Food Science and Engineering, Agricultural College Yanbian University Yanji China
| | - Muyao Wang
- Food Processing and Safety, Agricultural College Yanbian University Yanji China
| | - Mei Wang
- Dalian Academy of Agricultural Sciences Dalian China
| | - Zhanwen Qi
- Yanbian Han Gongfang Health Products Co., Ltd. Yanji China
| | - Chengbi Cui
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education Yanbian University Yanji China
- Food Science and Engineering, Agricultural College Yanbian University Yanji China
- Food Processing and Safety, Agricultural College Yanbian University Yanji China
| |
Collapse
|
9
|
Hugan Buzure Induces Autophagy and Apoptosis in Hepatocellular Carcinoma by Inhibiting PI3K/Akt/mTOR Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022. [DOI: 10.1155/2022/1618491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study explored the effects of Hugan Buzure (HBR) on cell apoptosis and autophagy in hepatocellular carcinoma (HCC) and the molecular mechanisms of the PI3K/Akt/mTOR signaling pathway. HepG2 and Huh7 cell viability was detected by the tetramethylazolium salt colorimetric (MTT) method. Cell proliferation was measured using the colony formation method. Hoechst 33258 staining and flow cytometry were employed to detect apoptosis. In addition, immunofluorescence was carried out to evaluate the expression of LC3. Western blot was performed to detect the expression of Bcl-2, Bax, Caspase-3, LC3, Beclin1, p62 (SQSTM1), and PI3K/Akt/mTOR signal pathway-related proteins in HCC cells. This work verified that HBR reduced HepG2 and Huh7 cell proliferation in a concentration-dependent manner. Treatment with HBR caused an obvious improvement of the apoptosis rate, accompanied by the increase in Bax/Bcl2, Caspase3, LC3II, and Beclin1 levels, respectively. Furthermore, HBR downregulated the expression of p62, p-PI3K, p-Akt, and p-mTOR proteins. HBR combined with HCQ enhanced HBR-induced apoptosis. In conclusion, HBR induced autophagy and apoptosis through PI3K/Akt/mTOR signaling pathway, leading to HCC cell death. This research preliminarily suggested the potential role of HBR in the treatment of HCC.
Collapse
|
10
|
Li J, Huang T, Lu J, Xu X, Zhang W. Metabonomic profiling of clubroot-susceptible and clubroot-resistant radish and the assessment of disease-resistant metabolites. FRONTIERS IN PLANT SCIENCE 2022; 13:1037633. [PMID: 36570889 PMCID: PMC9772615 DOI: 10.3389/fpls.2022.1037633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/16/2022] [Indexed: 06/17/2023]
Abstract
Plasmodiophora brassicae causes a serious threat to cruciferous plants including radish (Raphanus sativus L.). Knowledge on the pathogenic regularity and molecular mechanism of P. brassicae and radish is limited, especially on the metabolism level. In the present study, clubroot-susceptible and clubroot-resistant cultivars were inoculated with P. brassicae Race 4, root hairs initial infection of resting spores (107 CFU/mL) at 24 h post-inoculation and root galls symptom arising at cortex splitting stage were identified on both cultivars. Root samples of cortex splitting stage of two cultivars were collected and used for untargeted metabonomic analysis. We demonstrated changes in metabolite regulation and pathways during the cortex splitting stage of diseased roots between clubroot-susceptible and clubroot-resistant cultivars using untargeted metabonomic analysis. We identified a larger number of differentially regulated metabolites and heavier metabolite profile changes in the susceptible cultivar than in the resistant counterpart. The metabolites that were differentially regulated in both cultivars were mostly lipids and lipid-like molecules. Significantly regulated metabolites and pathways according to the P value and variable important in projection score were identified. Moreover, four compounds, including ethyl α-D-thioglucopyranoside, imipenem, ginsenoside Rg1, and 6-gingerol, were selected, and their anti-P. brassicae ability and effects on seedling growth were verified on the susceptible cultivar. Except for ethyl α-D-thioglucopyranoside, the remaining could inhibit clubroot development of varing degree. The use of 5 mg/L ginsenoside Rg1 + 5 mg/L 6-gingerol resulted in the lowest disease incidence and disease index among all treatments and enhanced seedling growth. The regulation of pathways or metabolites of carbapenem and ginsenoside was further explored. The results provide a preliminary understanding of the interaction between radish and P. brassicae at the metabolism level, as well as the development of measures for preventing clubroot.
Collapse
Affiliation(s)
- Jingwei Li
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Tingmin Huang
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Jinbiao Lu
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Xiuhong Xu
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Wanping Zhang
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| |
Collapse
|
11
|
Wen Z, Zhu H, Wu B, Zhang A, Wang H, Cheng Y, Zhao H, Li J, Liu M, Gao J. Cathepsin B plays a role in spermatogenesis and sperm maturation through regulating autophagy and apoptosis in mice. PeerJ 2022; 10:e14472. [PMID: 36518274 PMCID: PMC9744162 DOI: 10.7717/peerj.14472] [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: 08/09/2022] [Accepted: 11/06/2022] [Indexed: 12/03/2022] Open
Abstract
Spermatogenesis and sperm maturation are complex and highly ordered biological processes. Any failure or disorder in these processes can cause defects in sperm morphology, motility, and fertilization ability. Cathepsin B (CTSB) is involved in the regulation of a variety of pathological processes. In the present study, we found that CTSB was abundantly expressed in the male reproductive system, however, the specific role of CTSB in regulating spermatogenesis and sperm maturation remained elusive. Hence, we generated Ctsb -/- mice using CRISPR/Cas9 technology. In Ctsb -/- mice, sperm count was significantly decreased while the level of morphologically abnormal sperm was markedly increased. Additionally, these mice had significantly lower levels of progressive motility sperm and elevated levels of immobilized sperm. Histological analysis showed slight vacuolization in the testis epithelium, as well as the loss of epididymal epithelium cells. Further investigation showed that autophagic activity was inhibited and apoptotic activity was increased in both the testis and epididymis of Ctsb -/- mice. Together, our findings demonstrate that CTSB plays an important role in spermatogenesis and sperm maturation in mice.
Collapse
Affiliation(s)
- Zongzhuang Wen
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Haixia Zhu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Bin Wu
- Department of Reproductive Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Aizhen Zhang
- Department of Reproductive Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hongxiang Wang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Yin Cheng
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Hui Zhao
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Jianyuan Li
- Key Laboratory of Male Reproductive Health, Institute of Science and Technology, National Health Commission, Beijing, China
| | - Min Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Jiangang Gao
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China,School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| |
Collapse
|
12
|
Abstract
As a steroid skeleton-based saponin, ginsenoside Rh2 (G-Rh2) is one of the major bioactive ginsenosides from the plants of genus Panax L. Many studies have reported the notable pharmacological activities of G-Rh2 such as anticancer, antiinflammatory, antiviral, antiallergic, antidiabetic, and anti-Alzheimer's activities. Numerous preclinical studies have demonstrated the great potential of G-Rh2 in the treatment of a wide range of carcinomatous diseases in vitro and in vivo. G-Rh2 is able to inhibit proliferation, induce apoptosis and cell cycle arrest, retard metastasis, promote differentiation, enhance chemotherapy and reverse multi-drug resistance against multiple tumor cells. The present review mainly summarizes the anticancer effects and related mechanisms of G-Rh2 in various models as well as the recent advances in G-Rh2 delivery systems and structural modification to ameliorate its anticancer activity and pharmacokinetics characteristics.
Collapse
|
13
|
Jeong G, Shin SH, Kim SN, Na Y, Park BC, Cho JH, Park WS, Kim HJ. Ginsenoside Re prevents 3-methyladenine-induced catagen phase acceleration by regulating Wnt/β-catenin signaling in human dermal papilla cells. J Ginseng Res 2022; 47:440-447. [DOI: 10.1016/j.jgr.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/07/2022] [Accepted: 11/03/2022] [Indexed: 11/13/2022] Open
|
14
|
Autophagy in Hematological Malignancies. Cancers (Basel) 2022; 14:cancers14205072. [PMID: 36291856 PMCID: PMC9600546 DOI: 10.3390/cancers14205072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Autophagy is a dynamic and tightly regulated process that seems to have dual effects in cancer. In some contexts, it can induce carcinogenesis and promote cancer cell survival, whereas in others, it acts preventing tumor cell growth and tumor progression. Thus, autophagy functions seem to strictly depend on cancer ontogenesis, progression, and type. Here, we will dive into the current knowledge of autophagy in hematological malignancies and will highlight the main genetic components involved in each cancer type. Abstract Autophagy is a highly conserved metabolic pathway via which unwanted intracellular materials, such as unfolded proteins or damaged organelles, are digested. It is activated in response to conditions of oxidative stress or starvation, and is essential for the maintenance of cellular homeostasis and other vital functions, such as differentiation, cell death, and the cell cycle. Therefore, autophagy plays an important role in the initiation and progression of tumors, including hematological malignancies, where damaged autophagy during hematopoiesis can cause malignant transformation and increase cell proliferation. Over the last decade, the importance of autophagy in response to standard pharmacological treatment of hematological tumors has been observed, revealing completely opposite roles depending on the tumor type and stage. Thus, autophagy can promote tumor survival by attenuating the cellular damage caused by drugs and/or stabilizing oncogenic proteins, but can also have an antitumoral effect due to autophagic cell death. Therefore, autophagy-based strategies must depend on the context to create specific and safe combination therapies that could contribute to improved clinical outcomes. In this review, we describe the process of autophagy and its role on hematopoiesis, and we highlight recent research investigating its role as a potential therapeutic target in hematological malignancies. The findings suggest that genetic variants within autophagy-related genes modulate the risk of developing hemopathies, as well as patient survival.
Collapse
|
15
|
Ni B, Song X, Shi B, Wang J, Sun Q, Wang X, Xu M, Cao L, Zhu G, Li J. Research progress of ginseng in the treatment of gastrointestinal cancers. Front Pharmacol 2022; 13:1036498. [PMID: 36313365 PMCID: PMC9603756 DOI: 10.3389/fphar.2022.1036498] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/03/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer has become one of the major causes of human death. Several anticancer drugs are available; howeve their use and efficacy are limited by the toxic side effects and drug resistance caused by their continuous application. Many natural products have antitumor effects with low toxicity and fewer adverse effects. Moreover, they play an important role in enhancing the cytotoxicity of chemotherapeutic agents, reducing toxic side effects, and reversing chemoresistance. Consequently, natural drugs are being applied as potential therapeutic options in the field of antitumor treatment. As natural medicinal plants, some components of ginseng have been shown to have excellent efficacy and a good safety profile for cancer treatment. The pharmacological activities and possible mechanisms of action of ginseng have been identified. Its broad range of pharmacological activities includes antitumor, antibacterial, anti-inflammatory, antioxidant, anti-stress, anti-fibrotic, central nervous system modulating, cardioprotective, and immune-enhancing effects. Numerous studies have also shown that throuth multiple pathways, ginseng and its active ingredients exert antitumor effects on gastrointestinal (GI) tract tumors, such as esophageal, gastric, colorectal, liver, and pancreatic cancers. Herein, we introduced the main components of ginseng, including ginsenosides, polysaccharides, and sterols, etc., and reviewed the mechanism of action and research progress of ginseng in the treatment of various GI tumors. Futhermore, the pathways of action of the main components of ginseng are discussed in depth to promote the clinical development and application of ginseng in the field of anti-GI tumors.
Collapse
Affiliation(s)
- Baoyi Ni
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaotong Song
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bolun Shi
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia Wang
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Qianhui Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinmiao Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Manman Xu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luchang Cao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | | | - Jie Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jie Li,
| |
Collapse
|
16
|
Yin J, Zhuang J, Zhang X, Xu C, Lv S. Ginseng of different ages is affected by the accumulation of heavy metals in ginseng soil. PLoS One 2022; 17:e0269238. [PMID: 35696360 PMCID: PMC9191705 DOI: 10.1371/journal.pone.0269238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 05/17/2022] [Indexed: 11/18/2022] Open
Abstract
Heavy-metal pollution has been established to affect ginseng quality. However, this effect is still unknown in ginseng of different ages, emphasizing the need to investigate the effects of heavy metals in soils on ginseng growth. Herein, we determined the content of heavy metals (Cu, Cd, Pb, Hg, and As) in ginseng of different ages (2 to 6-year-old) and the corresponding soil samples. Then, the total ginsenosides content of ginseng and rate-limiting enzyme (HMGR, SQE, CYP450) activity in the synthesis of ginsenosides were assessed. Results from 200 differently-aged Chinese ginseng showed that increased ginsenoside content in 3 to 5-year-old ginseng was paralleled by increased heavy metal element content in ginseng and its soil. The activity of rate-limiting enzymes increased in the first four years of ginseng growth and then exhibited a steady or downward trend. Further analysis suggested that heavy metal elements in soils could directly affect ginsenoside content. Moreover, we found that Cu significantly affected the rate-limiting enzyme CYP450 activity. Further principal component analysis and correlation analysis found that heavy metals could obviously inhibit ginseng growth during the 5th and 6th years. Heavy metal content in soils has huge prospects for predicting ginsenoside, Cu and As content in ginseng. This study provided support for ginseng cultivation, quality research and quality assessment.
Collapse
Affiliation(s)
- Juxin Yin
- School of Information and Electrical Engineering, Zhejiang University City College, Hangzhou, People’s Republic of China
| | - Jianjian Zhuang
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Chaojian Xu
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Shaowu Lv
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| |
Collapse
|
17
|
Ginsenoside Rh2 inhibits breast cancer cell growth via ERβ-TNFα pathway. Acta Biochim Biophys Sin (Shanghai) 2022; 54:647-656. [PMID: 35593465 PMCID: PMC9828196 DOI: 10.3724/abbs.2022039] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Ginsenoside Rh2 is one of rare panaxidiols extracted from Panax ginseng and a potential estrogen receptor ligand that exhibits moderate estrogenic activity. However, the effect of Rh2 on growth inhibition and its underlying molecular mechanism in human breast cells are not fully understood. In this study, we tested cell viability by MTT and colony formation assays. Cell growth and cell cycle were determined to investigate the effect of ginsenoside Rh2 by flow cytometry. The expressions of estrogen receptors (ERs), TNFα, and apoptosis-related proteins were detected by qPCR and western blot analysis. The mechanisms of ERα and ERβ action were determined using transfection and inhibitors. Antitumor effect of ginsenoside Rh2 against MCF-7 cells was investigated in xenograft mice. Our results showed that ginsenoside Rh2 induced apoptosis and G1/S phase arrest in MCF-7 cells. Treatment of cells with ginsenoside Rh2 down-regulated protein levels of ERα, and up-regulated mRNA and protein levels of ERβ and TNFα. We also found that ginsenoside Rh2-induced TNFα over-expression is through up-regulation of ERβ initiated by ginsenoside Rh2. Furthermore, ginsenoside Rh2 induced MCF-7 cell apoptosis via estrogen receptor β-TNFα pathway in vivo. These results demonstrate that ginsenoside Rh2 promotes TNFα-induced apoptosis and G1/S phase arrest via regulation of ERβ.
Collapse
|
18
|
Patangrao Renushe A, Kumar Banothu A, Kumar Bharani K, Mekala L, Mahesh Kumar J, Neeradi D, Durga Veera Hanuman D, Gadige A, Khurana A. Vincamine, an active constituent of Vinca rosea ameliorates experimentally induced acute lung injury in Swiss albino mice through modulation of Nrf-2/NF-κB signaling cascade. Int Immunopharmacol 2022; 108:108773. [PMID: 35453074 DOI: 10.1016/j.intimp.2022.108773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 11/12/2022]
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is one of the leading pulmonary inflammatory disorders causing significant morbidity and mortality. Vincamine is a novel phytochemical with promising anti-inflammatory properties. In the current work, the protective effect of vincamine was studied in vitro (Raw 264.7 macrophages) and in vivo against lipopolysaccharide (LPS) induced ALI in Swiss albino mice. Vincamine significantly reduced nitrite and TNF-α release from the LPS stimulated macrophages and increased the levels of IL-10, indicating potent anti-inflammatory effects. It was observed that vincamine at the dose of 40 mg/kg, significantly reduced LPS induced inflammatory cell count in blood and in bronchoalveolar lavage (BAL) fluid. Further, vincamine exerted potent suppression of inflammation by reducing the expression of proinflammatory cytokines, while significantly increased (p < 0.001) the expression of anti-inflammatory cytokine (IL-10 and IL-22). Interestingly, histological changes were reversed in vincamine treated groups in a dose-dependent manner. Immunohistochemical analysis revealed significantly enhanced expression of NF-κB, TNF-α and COX-2 while reduced expression of Nrf-2 in disease control group, which were significantly (p < 0.001) ameliorated by vincamine. We, to the best of our knowledge, report for the first time that vincamine possesses protective potential against LPS induced inflammation and oxidative stress, possibly by inhibiting the NF-κB cascade, while positively regulating the Nrf-2 pathway. These findings are of potential relevance for COVID-19 management concerning the fact that lung injury and ARDS are its critical features.
Collapse
Affiliation(s)
- Akshata Patangrao Renushe
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad - 500030, Telangana, India
| | - Anil Kumar Banothu
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad - 500030, Telangana, India.
| | - Kala Kumar Bharani
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Warangal - 506166, Telangana, India
| | - Lakshman Mekala
- Department of Veterinary Pathology, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad - 500030, Telangana, India
| | - Jerald Mahesh Kumar
- Animal House, Council for Scientific and Industrial Research (CSIR) - Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad - 500007, Telangana, India
| | - Dinesh Neeradi
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad - 500030, Telangana, India
| | - Donga Durga Veera Hanuman
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad - 500030, Telangana, India
| | - Ambica Gadige
- Department of Veterinary Medicine, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad - 500030, Telangana, India
| | - Amit Khurana
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad - 500030, Telangana, India; Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Warangal - 506166, Telangana, India; Centre for Biomedical Engineering (CBME), Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi - 110016, India.
| |
Collapse
|
19
|
He X, Liao Y, Liu J, Sun S. Research Progress of Natural Small-Molecule Compounds Related to Tumor Differentiation. Molecules 2022; 27:2128. [PMID: 35408534 PMCID: PMC9000768 DOI: 10.3390/molecules27072128] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 11/25/2022] Open
Abstract
Tumor differentiation is a therapeutic strategy aimed at reactivating the endogenous differentiation program of cancer cells and inducing cancer cells to mature and differentiate into other types of cells. It has been found that a variety of natural small-molecule drugs can induce tumor cell differentiation both in vitro and in vivo. Relevant molecules involved in the differentiation process may be potential therapeutic targets for tumor cells. Compared with synthetic drugs, natural small-molecule antitumor compounds have the characteristics of wide sources, structural diversity and low toxicity. In addition, natural drugs with structural modification and transformation have relatively concentrated targets and enhanced efficacy. Therefore, using natural small-molecule compounds to induce malignant cell differentiation represents a more targeted and potential low-toxicity means of tumor treatment. In this review, we focus on natural small-molecule compounds that induce differentiation of myeloid leukemia cells, osteoblasts and other malignant cells into functional cells by regulating signaling pathways and the expression of specific genes. We provide a reference for the subsequent development of natural small molecules for antitumor applications and promote the development of differentiation therapy.
Collapse
Affiliation(s)
- Xiaoli He
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Yongkang Liao
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Shuming Sun
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
20
|
Chen YY, Liang JJ, Wang DL, Chen JB, Cao JP, Wang Y, Sun CD. Nobiletin as a chemopreventive natural product against cancer, a comprehensive review. Crit Rev Food Sci Nutr 2022; 63:6309-6329. [PMID: 35089821 DOI: 10.1080/10408398.2022.2030297] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As a leading cause of death, second only to heart disease, cancer has always been one of the burning topics in medical research. When targeting multiple signal pathways in tumorigenesis chemoprevention, using natural or synthetic anti-cancer drugs is a vital strategy to reduce cancer damage. However, toxic effects, multidrug resistance (MDR) as well as cancer stem cells (CSCs) all prominently limited the clinical application of conventional anticancer drugs. With low side effects, strong biological activity, unique mechanism, and wide range of targets, natural products derived from plants are considered significant sources for new drug development. Nobiletin is one of the most attractive compounds, a unique flavonoid primarily isolated from the peel of citrus fruits. Numerous studies in vitro and in vivo have suggested that nobiletin and its derivatives possess the eminent potential to become effective cancer chemoprevention agents through various cellular and molecular levels. This article aims to comprehensively review the anticancer efficacy and specific mechanisms of nobiletin, enhancing our understanding of its chemoprevention properties and providing the latest research findings. At the end of this review, we also give some discussion and future perspectives regarding the challenges and opportunities in nobiletin efficient exploitation.
Collapse
Affiliation(s)
- Yun-Yi Chen
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Jiao-Jiao Liang
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Deng-Liang Wang
- Citrus Research Institute, Quzhou Academy of Agricultural Sciences, Quzhou, China
| | - Jie-Biao Chen
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Jin-Ping Cao
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Yue Wang
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| | - Chong-De Sun
- Laboratory of Fruit Quality Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, China
| |
Collapse
|
21
|
Research Advances in Antitumor Mechanism of Evodiamine. J CHEM-NY 2022. [DOI: 10.1155/2022/2784257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Evodiamine is a natural alkaloid extracted from Fructus Evodia. This bioactive alkaloid has been reported to have a wide range of biological activities, including anti-injury, antiobesity, vasodilator, and anti-inflammatory effects. In recent years, it has been found that evodiamine has tumor-suppressive effects on a variety of tumors. There is growing evidence that evodiamine can inhibit the rapid proliferation of tumor cells, induce cell cycle arrest at a certain phase, increase the incidence of apoptosis, promote autophagy, inhibit microangiogenesis and migration, and regulate immunotherapy. Evodiamine can inhibit Wnt/β-catenin, mTOR, NF-κB, PI3K/AKT, JAK-STAT, and other signaling pathways in various cancer cells, and it can significantly downregulate the expression of many tumor markers, such as VEGF and COX-2. These facts partially explain the antitumor mechanism of evodiamine. In this article, the antitumor mechanism of evodiamine was reviewed to provide the basis for its clinical application and therapeutic development in the future.
Collapse
|
22
|
Lai J, Tang Y, Yang F, Chen J, Huang FH, Yang J, Wang L, Qin D, Law BYK, Wu AG, Wu JM. Targeting autophagy in ethnomedicine against human diseases. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114516. [PMID: 34487846 DOI: 10.1016/j.jep.2021.114516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/29/2021] [Accepted: 08/09/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In the past five years, ethnopharmacy-based drugs have been increasingly used in clinical practice. It has been reported that hundreds of ethnopharmacy-based drugs can modulate autophagy to regulate physiological and pathological processes, and ethnomedicines also have certain therapeutic effects on illnesses, revealing the important roles of these medicines in regulating autophagy and treating diseases. AIM OF THE STUDY This study reviews the regulatory effects of natural products on autophagy in recent years, and discusses their pharmacological effects and clinical applications in the process of diseases. It provides a preliminary literature basis and reference for the research of plant drugs in the regulation of autophagy. MATERIALS AND METHODS A comprehensive systematic review in the fields of relationship between autophagy and ethnomedicine in treating diseases from PubMed electronic database was performed. Information was obtained from documentary sources. RESULTS We recorded some illnesses associated with autophagy, then classified them into different categories reasonably. Based on the uses of these substances in different researches of diseases, a total of 80 active ingredients or compound preparations of natural drugs were searched. The autophagy mechanisms of these substances in the treatments of divers diseases have been summarized for the first time, we also looked forward to the clinical application of some of them. CONCLUSIONS Autophagy plays a key function in lots of illnesses, the regulation of autophagy has become one of the important means to prevent and treat these diseases. About 80 compounds and preparations involved in this review have been proved to have therapeutic effects on related diseases through the mechanism of autophagy. Experiments in vivo and in vitro showed that these compounds and preparations could treat these diseases by regulating autophagy. The typical natural products curcumin and tripterine have powerful roles in regulating autophagy and show good and diversified curative effects.
Collapse
Affiliation(s)
- Jia Lai
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yong Tang
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Fei Yang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Fei-Hong Huang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China
| | - Jing Yang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China
| | - Dalian Qin
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - An-Guo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China.
| | - Jian-Ming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
23
|
Bian S, Liu M, Yang S, Lu S, Wang S, Bai X, Zhao D, Wang J. 20(S)-Ginsenoside Rh2-induced apoptosis and protective autophagy in cervical cancer cells by inhibiting AMPK/mTOR pathway. Biosci Biotechnol Biochem 2021; 86:92-103. [PMID: 34718401 DOI: 10.1093/bbb/zbab189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/25/2021] [Indexed: 01/30/2023]
Abstract
20(S)-Ginsenoside Rh2 (GRh2) has various biological activities including anticancer effects. However, no reports have investigated the connection between autophagy and apoptosis in HeLa cells treated with 20(S)-GRh2. In this study, we found that 20(S)-GRh2 suppressed proliferation and induced apoptosis in HeLa cells by activating the intrinsic apoptotic pathway and causing mitochondrial dysfunction. 20(S)-GRh2 enhanced cell autophagy through promoting the phosphorylation of AMPK, depressed the phosphorylation of AKT, and suppressed mTOR activity. Furthermore, treatment with the autophagy inhibitor 3-methyladenine (3-MA) enhanced 20(S)-GRh2-induced apoptosis, while the autophagy inducer rapamycin promoted cell survival. Moreover, the apoptosis inhibitor Z-VAD-FMK significantly restrained the apoptosis and autophagy induced by 20(S)-GRh2 in HeLa cells. We found that 20(S)-ginsenoside Rh2-induced protective autophagy promotes apoptosis of cervical cancer cells by inhibiting AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Shuai Bian
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meichen Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Song Yang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Shuyan Lu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Siming Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xueyuan Bai
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jiawen Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
24
|
Liu Y, Yu S, Xing X, Qiao J, Yin Y, Wang J, Liu M, Zhang W. Ginsenoside Rh2 stimulates the production of mitochondrial reactive oxygen species and induces apoptosis of cervical cancer cells by inhibiting mitochondrial electron transfer chain complex. Mol Med Rep 2021; 24:873. [PMID: 34713297 PMCID: PMC8569524 DOI: 10.3892/mmr.2021.12513] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/07/2021] [Indexed: 11/29/2022] Open
Abstract
Ginsenoside Rh2 (G-Rh2) is a monomeric compound that extracted from ginseng and possesses anti-cancer activities both in vitro and in vivo. Previously, we reported that G-Rh2 induces apoptosis in HeLa cervical cancer cells and that the process was related to reactive oxygen species (ROS) accumulation and mitochondrial dysfunction. However, the upstream mechanisms of G-Rh2, along with its cellular targets, remain to be elucidated. In the present study, the Cell Counting Kit-8 assay, flow cytometry and Hoechst staining revealed that G-Rh2 significantly inhibited cell viability and induced apoptosis of cervical cancer cells. However, G-Rh2 was demonstrated to be non-toxic to End1/e6e7 cells. JC-1, rhodamine 123 staining, oxidative phosphorylation and glycolysis capacity assays demonstrated that G-Rh2 exposure caused an immediate decrease in mitochondrial transmembrane potential due to its inhibition of mitochondrial oxidative phosphorylation, as well as glycolysis, both of which reduced cellular ATP production. Western blotting and electron transport chain (ETC) activity assays revealed that G-Rh2 significantly inhibited the activity of ETC complexes I, III and V. Overexpression of ETC complex III partially significantly restored mitochondrial ROS and inhibited the apoptosis of cervical cancer cells induced by G-Rh2. The predicted results of binding energy in molecular docking, confirmed that G-Rh2 was highly likely to induce mitochondrial ROS production and promote cell apoptosis by targeting the ETC complex, especially for ETC complex III. Taken together, the present results revealed the potential anti-cervical cancer activity of G-Rh2 and provide direct evidence for the contribution of impaired ETC complex activity to cervical cancer cell death.
Collapse
Affiliation(s)
- Ying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Shiting Yu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Xin Xing
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Juhui Qiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yiqiu Yin
- GeneScience Pharmaceuticals Co., Ltd., Changchun, Jilin 130012, P.R. China
| | - Jiawen Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Meichen Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Wei Zhang
- Scientific Research Department, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| |
Collapse
|
25
|
Ginsenoside Rh2 Suppresses Metastasis and Growth of Colon Cancer via miR-491. JOURNAL OF ONCOLOGY 2021; 2021:6815713. [PMID: 34603449 PMCID: PMC8486518 DOI: 10.1155/2021/6815713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/20/2023]
Abstract
Ginsenoside Rh2 is considered as a new direction for future cancer treatment because of its excellent anticancer effect. However, due to its low bioavailability, it cannot exert its significant anticancer effect when applied directly to the human body. Chitosan (CS), a nanomaterial, has been verified to be able to enhance drug efficacy via its coating for drugs. Thus, we designed this study to investigate the impact of CS-coated ginsenoside Rh2 on the metastasis and growth of colon cancer (CC). First, ginsenoside Rh2 chitosan tripolyphosphate (CS-Rh2-TPP) nanoparticles (NPs) were constructed, and MTT, transwell, scratch adhesion, and flow cytometry assays were carried out for determining the impact of CS-Rh2-TPP at various concentrations on growth, metastasis, and apoptosis of colon cancer cells (CCCs). qRT-PCR was used to detect the expression of mircoRNA-491 (miR-491) in CCCs. According to TEM-based image analysis, CS-Rh2-TPP NPs were spherical or spheroidal in even distribution, with a particle size of about 220 mm and a zeta potential of −44.58 ± 2.84 mV. Additionally, CCCs presented lower miR-491 than normal colon cells, and its relative expression in CCCs showed a stronger increase after intervention of CS-Rh2-TPP than that after intervention of ginsenoside Rh2. Moreover, CS-Rh2-TPP suppressed the activity, invasion, as well as migration of CCCs and accelerated their apoptosis more significantly than ginsenoside Rh2. According to these results, CS-Rh2-TPP is able to upregulate miR-491 in CCCs, thus suppressing the metastasis and growth of CC.
Collapse
|
26
|
Xia X, Tao J, Ji Z, Long C, Hu Y, Zhao Z. Increased antitumor efficacy of ginsenoside Rh 2 via mixed micelles: in vivo and in vitro evaluation. Drug Deliv 2021; 27:1369-1377. [PMID: 32998576 PMCID: PMC7580790 DOI: 10.1080/10717544.2020.1825542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The aim of this work is to apply Solutol® HS15 and TPGS to prepare self-assembled micelles loading with ginsenoside Rh2 to increase the solubility of ginsenoside Rh2, hence, improving the antitumor efficacy. Ginsenoside Rh2-mixed micelles (Rh2-M) were prepared by thin film dispersion method. The optimal Rh2-M was characterized by particle size, morphology, and drug encapsulation efficiency. The enhancement of in vivo anti-tumor efficacy of Rh2-M was evaluated by nude mice bearing tumor model. The solubility of Rh2 in self-assembled micelles was increased approximately 150-folds compared to free Rh2. In vitro results demonstrated that the particle size of Rh2-M is 74.72 ± 2.63 nm(PDI = 0.147 ± 0.15), and the morphology of Rh2-M is spherical or spheroid, and the EE% and LE% are 95.27 ± 1.26% and 7.68 ± 1.34%, respectively. The results of in vitro cell uptake and in vivo imaging showed that Rh2-M could not only increase the cell uptake of drugs, but also transport drug to tumor sites, prolonging the retention time. In vitro cytotoxicity and in vivo antitumor results showed that the anti-tumor effect of Rh2 can be effectively improved by Rh2-M. Therefore, Solutol® HS15 and TPGS could be used to entrapping Rh2 into micelles, enhancing solubility and antitumor efficacy.
Collapse
Affiliation(s)
- Xiaojing Xia
- Department of Pharmaceutics, Zhejiang Pharmaceutical College, Ningbo, PR China
| | - Jin Tao
- Department of Pharmaceutics, Zhejiang Pharmaceutical College, Ningbo, PR China
| | - Zhuwa Ji
- Department of Pharmaceutics, Zhejiang Pharmaceutical College, Ningbo, PR China
| | - Chencheng Long
- Department of Pharmaceutics, Zhejiang Pharmaceutical College, Ningbo, PR China
| | - Ying Hu
- Department of Pharmaceutics, Zhejiang Pharmaceutical College, Ningbo, PR China
| | - Zhiying Zhao
- Department of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, PR China
| |
Collapse
|
27
|
Bian S, Zhao Y, Li F, Lu S, He Z, Wang S, Bai X, Zhao D, Liu M, Wang J. Total ginsenosides induce autophagic cell death in cervical cancer cells accompanied by downregulation of bone marrow stromal antigen-2. Exp Ther Med 2021; 22:667. [PMID: 33986832 PMCID: PMC8112150 DOI: 10.3892/etm.2021.10099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 03/15/2021] [Indexed: 12/25/2022] Open
Abstract
Ginsenosides are important active components in Panax ginseng. In the present study, total ginsenosides (TGNs) were demonstrated to enhance autophagy by promoting acidic vacuole organelle formation, recruitment of enhanced green fluorescent protein-microtubule-associated protein light chain 3 and expression of autophagy-related factors in cervical cancer cell lines. TGN markedly increased the expression of p62 at the transcriptional level, but decreased p62 protein expression in the presence of actinomycin D. The autophagic regulatory effect was reversible. TGN (≤120 µg/ml) did not affect the proliferation of cervical cancer cells under normal culture conditions, but markedly inhibited the growth of serum-deprived cells. Treatment with an inhibitor of autophagy (3-methyladenine) impaired TGN-induced cell death. This suggested that TGN caused autophagic cell death. In addition, western blot analysis demonstrated that the protein level of bone marrow stromal antigen-2 (BST-2) was downregulated by TGN. Upregulation of BST-2 reduced cell death. The results of the combined actions of various monomeric ginsenosides in TGN provide the molecular basis to develop TGN as a promising candidate for cancer therapy.
Collapse
Affiliation(s)
- Shuai Bian
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yue Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Fangyu Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Shuyan Lu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Ziyan He
- College of Chemistry, Jilin University, Changchun, Jilin 13012, P.R. China
| | - Siming Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Xueyuan Bai
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Meichen Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Jiawen Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| |
Collapse
|
28
|
Paving the Road Toward Exploiting the Therapeutic Effects of Ginsenosides: An Emphasis on Autophagy and Endoplasmic Reticulum Stress. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1308:137-160. [PMID: 33861443 DOI: 10.1007/978-3-030-64872-5_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Programmed cell death processes such as apoptosis and autophagy strongly contribute to the onset and progression of cancer. Along with these lines, modulation of cell death mechanisms to combat cancer cells and elimination of resistance to apoptosis is of great interest. It appears that modulation of autophagy and endoplasmic reticulum (ER) stress with specific agents would be beneficial in the treatment of several disorders. Interestingly, it has been suggested that herbal natural products may be suitable candidates for the modulation of these processes due to few side effects and significant therapeutic potential. Ginsenosides are derivatives of ginseng and exert modulatory effects on the molecular mechanisms associated with autophagy and ER stress. Ginsenosides act as smart phytochemicals that confer their effects by up-regulating ATG proteins and converting LC3-I to -II, which results in maturation of autophagosomes. Not only do ginsenosides promote autophagy but they also possess protective and therapeutic properties due to their capacity to modulate ER stress and up- and down-regulate and/or dephosphorylate UPR transducers such as IRE1, PERK, and ATF6. Thus, it would appear that ginsenosides are promising agents to potentially restore tissue malfunction and possibly eliminate cancer.
Collapse
|
29
|
He XL, Xu XH, Shi JJ, Huang M, Wang Y, Chen X, Lu JJ. Anticancer Effects of Ginsenoside Rh2: A Systematic Review. Curr Mol Pharmacol 2021; 15:179-189. [PMID: 33687905 DOI: 10.2174/1874467214666210309115105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/22/2020] [Accepted: 01/18/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND As one of the effective pharmacological constituents of Ginseng Radix et Rhizoma, ginsenoside Rh2 (Rh2) exerts a remarkable anticancer effect on various cancer cell lines in vitro and strongly inhibits tumor growth in vivo without severe toxicity. OBJECTIVE This article reviewed existing evidence supporting the anticancer effects of Rh2 to classify and conclude previous and current knowledge on the mechanisms and therapeutic effects of Rh2, as well as to promote the clinical application of this natural product. CONCLUSION This article reviewed the anticancer efficacies and mechanisms of Rh2, including the induction of cell cycle arrest and programmed cell death, repression of metastasis, alleviation of drug resistance, and regulation of the immune system. Finally, this paper discussed the research and application prospects of Rh2.
Collapse
Affiliation(s)
- Xin-Ling He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Xiao-Huang Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Jia-Jie Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Mingqing Huang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122. China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| |
Collapse
|
30
|
Wang J, Bian S, Wang S, Yang S, Zhang W, Zhao D, Liu M, Bai X. Ginsenoside Rh2 represses autophagy to promote cervical cancer cell apoptosis during starvation. Chin Med 2020; 15:118. [PMID: 33292331 PMCID: PMC7661217 DOI: 10.1186/s13020-020-00396-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022] Open
Abstract
Background Cancer cells through autophagy-mediated recycling to meet the metabolic demands of growth and proliferation. The steroidal saponin 20(S)-ginsenoside Rh2 effectively inhibits the growth and survival of a variety of tumor cell lines and animal models, but the effects of Rh2 on autophagy remain elusive. Methods Cell viability was measured by CCK-8 (cell counting kit-8) assays. Apoptosis, ROS generation and mitochondrial membrane potential were analyzed by flow cytometry. Western blot analyses were used to determine changes in protein levels. Morphology of apoptotic cells and autophagosome accumulation were analyzed by DAPI staining and transmission electron microscopy. Autophagy induction was monitored by acidic vesicular organelle staining, EGFP-LC3 and mRFP-GFP-LC3 transfection. Atg7 siRNA and autophagy regulator was used to assess the effect of autophagy on apoptosis induced by G-Rh2. Results In this study, we found that low concentration G-Rh2 attenuated cancer cell growth and induced apoptosis upon serum-free starvation. Caspase 3 inhibitors failed to block apoptosis in G-Rh2-treated cells, indicating a caspase-independent mechanism. G-Rh2-treated cells in serum-deprived conditions showed impaired mitochondrial function, increased release and nuclear translocation of apoptosis-inducing factor, but little changes in the mitochondrial and cytoplasmic distributions of cytochrome C. Annexin A2 overexpression in 293T cells inhibited G-Rh2-induced apoptosis under serum-starved conditions. Meanwhile, G-Rh2 reduced lysosomal activity and inhibited the fusion of autophagosome and lysosome, leading to a block of autophagic flux. Knockdown Atg7 significantly inhibited autophagy and triggered AIF-induced apoptosis in serm free condition. The autophagy inducer significantly decreased the apoptosis levels of G-Rh2-treated cells in serum-free conditions. Conclusions Under nutrient deficient conditions, G-Rh2 represses autophagy in cervical cancer cells and enhanced apoptosis through an apoptosis-inducing factor mediated pathway.
Collapse
Affiliation(s)
- Jiawen Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Boshuo Road 1035, Changchun, 130117, Jilin, People's Republic of China
| | - Shuai Bian
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Boshuo Road 1035, Changchun, 130117, Jilin, People's Republic of China
| | - Siming Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Boshuo Road 1035, Changchun, 130117, Jilin, People's Republic of China
| | - Song Yang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Boshuo Road 1035, Changchun, 130117, Jilin, People's Republic of China
| | - Wanying Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Boshuo Road 1035, Changchun, 130117, Jilin, People's Republic of China
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Boshuo Road 1035, Changchun, 130117, Jilin, People's Republic of China
| | - Meichen Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Boshuo Road 1035, Changchun, 130117, Jilin, People's Republic of China.
| | - Xueyuan Bai
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Boshuo Road 1035, Changchun, 130117, Jilin, People's Republic of China.
| |
Collapse
|
31
|
Zhao Q, Peng C, Zheng C, He XH, Huang W, Han B. Recent Advances in Characterizing Natural Products that Regulate Autophagy. Anticancer Agents Med Chem 2020; 19:2177-2196. [PMID: 31749434 DOI: 10.2174/1871520619666191015104458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/16/2018] [Accepted: 08/26/2019] [Indexed: 02/07/2023]
Abstract
Autophagy, an intricate response to nutrient deprivation, pathogen infection, Endoplasmic Reticulum (ER)-stress and drugs, is crucial for the homeostatic maintenance in living cells. This highly regulated, multistep process has been involved in several diseases including cardiovascular and neurodegenerative diseases, especially in cancer. It can function as either a promoter or a suppressor in cancer, which underlines the potential utility as a therapeutic target. In recent years, increasing evidence has suggested that many natural products could modulate autophagy through diverse signaling pathways, either inducing or inhibiting. In this review, we briefly introduce autophagy and systematically describe several classes of natural products that implicated autophagy modulation. These compounds are of great interest for their potential activity against many types of cancer, such as ovarian, breast, cervical, pancreatic, and so on, hoping to provide valuable information for the development of cancer treatments based on autophagy.
Collapse
Affiliation(s)
- Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Chuan Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Xiang-Hong He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China.,The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, United States
| |
Collapse
|
32
|
Zare-Zardini H, Alemi A, Taheri-Kafrani A, Hosseini SA, Soltaninejad H, Hamidieh AA, Haghi Karamallah M, Farrokhifar M, Farrokhifar M. Assessment of a New Ginsenoside Rh2 Nanoniosomal Formulation for Enhanced Antitumor Efficacy on Prostate Cancer: An in vitro Study. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3315-3324. [PMID: 32884236 PMCID: PMC7431455 DOI: 10.2147/dddt.s261027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/18/2020] [Indexed: 12/14/2022]
Abstract
Introduction Ginsenoside Rh2, purified from the Panax ginseng root, has been demonstrated to possess anticancer properties against various cancerous cells including colorectal, breast, skin, ovarian, prostate, and liver cancerous cells. However, the poor bioavailability, low stability on gastrointestinal systems, and fast plasma elimination limit further clinical applications of Ginsenoside Rh2 for cancer treatments. In this study, a novel formulation of niosomal Ginsenoside Rh2 was prepared using the thin film hydration technique. Methods The niosomal formulation contained Span 60 and cholesterol, and cationic lipid DOTAP was evaluated by determining particle size distribution, encapsulation efficiency, the polydispersity index (PDI), and surface morphology. The cytotoxic effects of free Ginsenoside Rh2 and Ginsenoside Rh2-loaded niosomes were determined using the MTT method in the PC3 prostate cancer cell line. For the investigation of the in vitro cellular uptake of Ginsenoside Rh2-loaded niosome, two formulations were prepared: the Ginsenoside Rh2-loaded niosomal formula containing 5% DOTAP and the Ginsenoside Rh2-loaded niosomal formula without DOTAP. Results The mean size, DPI, zeta potential, and encapsulation efficiency of the Ginsenoside Rh2-loaded nanoniosomal formulation containing DOTAP were 93.5±2.1 nm, 0.203±0.01, +4.65±0.65, and 98.32% ±2.4, respectively. The niosomal vesicles were found to be round and have a smooth surface. The release profile of Ginsenoside Rh2 from niosome was biphasic. Furthermore, a two-fold reduction in the Ginsenoside Rh2 concentration was measured when Ginsenoside Rh2 was administered in a nanoniosomal form compared to free Ginsenoside Rh2 solutions in the PC3 prostate cancer cell line. After storage for 90 days, the encapsulation efficiency, vesicle size, PDI, and zeta potential of the optimized formulation did not significantly change compared to the freshly prepared samples. The cellular uptake experiments of the niosomal formulation demonstrated that by adding DOTAP to the niosomal formulation, the cellular uptake was enhanced. Discussion The enhanced cellular uptake and cytotoxic activity of the Ginsenoside Rh2 nanoniosomal formulation on the PC3 cell make it an attractive candidate for application as a nano-sized delivery vehicle to transfer Ginsenoside Rh2 to cancer cells.
Collapse
Affiliation(s)
- Hadi Zare-Zardini
- Hematology and Oncology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Sciences, Farhangian University, Isfahan, Iran.,Medical Nanotechnology &Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ashraf Alemi
- Abadan Faculty of Medical Sciences, Abadan, Iran
| | - Asghar Taheri-Kafrani
- Department of Biotechnology, Faculty of Advanced Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Seyed Ahmad Hosseini
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Soltaninejad
- Tissue Bank & Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Ali Hamidieh
- Stem Cell and Regenerative Medicine Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Majid Farrokhifar
- Department of Pediatrics, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | |
Collapse
|
33
|
Li X, Chu S, Lin M, Gao Y, Liu Y, Yang S, Zhou X, Zhang Y, Hu Y, Wang H, Chen N. Anticancer property of ginsenoside Rh2 from ginseng. Eur J Med Chem 2020; 203:112627. [PMID: 32702586 DOI: 10.1016/j.ejmech.2020.112627] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022]
Abstract
Ginseng has been used as a well-known traditional Chinese medicine since ancient times. Ginsenosides as its main active constituents possess a broad scope of pharmacological properties including stimulating immune function, enhancing cardiovascular health, increasing resistance to stress, improving memory and learning, developing social functioning and mental health in normal persons, and chemotherapy. Ginsenoside Rh2 (Rh2) is one of the major bioactive ginsenosides from Panax ginseng. When applied to cancer treatment, Rh2 not only exhibits the anti-proliferation, anti-invasion, anti-metastasis, induction of cell cycle arrest, promotion of differentiation, and reversal of multi-drug resistance activities against multiple tumor cells, but also alleviates the side effects after chemotherapy or radiotherapy. In the past decades, nearly 200 studies on Rh2 in the treatment of cancer have been published, however no specific reviews have been conducted by now. So the purpose of this review is to provide a systematic summary and analysis of the anticancer effects and the potential mechanisms of Rh2 extracted from Ginseng then give a future prospects about it. In the end of this paper the metabolism and derivatives of Rh2 also have been documented.
Collapse
Affiliation(s)
- Xun Li
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China; Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, PR China; Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Shifeng Chu
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Meiyu Lin
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Yan Gao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Yingjiao Liu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Songwei Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Xin Zhou
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Yani Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Yaomei Hu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Huiqin Wang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Naihong Chen
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China; Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, PR China; Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China.
| |
Collapse
|
34
|
Inamura A, Muraoka-Hirayama S, Sakurai K. Loss of Mitochondrial DNA by Gemcitabine Triggers Mitophagy and Cell Death. Biol Pharm Bull 2020; 42:1977-1987. [PMID: 31787713 DOI: 10.1248/bpb.b19-00312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gemcitabine (2,2-difluorodeoxycytidine nucleic acid), an anticancer drug exhibiting a potent ability to kill cancer cells, is a frontline chemotherapy drug. Although some chemotherapeutic medicines are known to induce nuclear DNA damage, no investigation into mitochondrial DNA (mtDNA) damage currently exists. When we treated insulinoma pancreatic β-cells (line INS-1) with high mitochondrial activity with gemcitabine for 24 h, the mtDNA contents were decreased. Gemcitabine induced a decrease in the number of mitochondria and the average potential of mitochondrial membrane in the cell but increased the superoxide anion radical levels. We observed that treatment with gemcitabine to induce cell death accompanied by autophagy-related protein markers, Atg5 and Atg7; these were significantly prevented by the autophagy inhibitors. The localization of Atg5 co-occurred with the location of mitochondria with membranes having high potential and mitophagy in cells treated with gemcitabine. The occurrence of mitophagy was inhibited by the inhibitors of the phosphatidylinositol 3-kinase/Akt pathway. Our results led us to the conclusion that gemcitabine induced cell death through mitophagy with the loss of mtDNA. These findings may provide a rationale for the combination of mtDNA damage with mitophagy in future clinical applications for cancer cells.
Collapse
Affiliation(s)
- Akihiro Inamura
- Division of Life Science, Department of Pharmacy, Hokkaido University of Science
| | | | - Koichi Sakurai
- Division of Life Science, Department of Pharmacy, Hokkaido University of Science
| |
Collapse
|
35
|
Wang M, Li H, Liu W, Cao H, Hu X, Gao X, Xu F, Li Z, Hua H, Li D. Dammarane-type leads panaxadiol and protopanaxadiol for drug discovery: Biological activity and structural modification. Eur J Med Chem 2020; 189:112087. [PMID: 32007667 DOI: 10.1016/j.ejmech.2020.112087] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/14/2019] [Accepted: 01/20/2020] [Indexed: 12/13/2022]
Abstract
Based on the definite therapeutic benefits, such as neuroprotective, cardioprotective, anticancer, anti-diabetic and so on, the Panax genus which contains many valuable plants, including ginseng (Panax ginseng C.A. Meyer), notoginseng (Panax notoginseng) and American ginseng (Panax quinquefolius L.), attracts research focus. Actually, the biological and pharmacological effects of the Panax genus are mainly attributed to the abundant ginsenosides. However, the low membrane permeability and the gastrointestinal tract influence seriously limit the absorption and bioavailability of ginsenosides. The acid or base hydrolysates of ginsenosides, 20 (R,S)-panaxadiol and 20 (R,S)-protopanaxadiol showed improved bioavailability and diverse pharmacological activities. Moreover, relative stable skeletons and active hydroxyl group at C-3 position and other reactive sites are suitable for structural modification to improve biological activities. In this review, the pharmacological activities of panaxadiol, protopanaxadiol and their structurally modified derivatives are comprehensively summarized.
Collapse
Affiliation(s)
- Mingying Wang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Haonan Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Hao Cao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Xu Hu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Xiang Gao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Zhanlin Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, And School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
36
|
Zhang R, Chen J, Mao L, Guo Y, Hao Y, Deng Y, Han X, Li Q, Liao W, Yuan M. Nobiletin Triggers Reactive Oxygen Species-Mediated Pyroptosis through Regulating Autophagy in Ovarian Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:1326-1336. [PMID: 31955565 DOI: 10.1021/acs.jafc.9b07908] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ovarian cancer is one of the most serious female malignancies worldwide. Despite intensive efforts being made to overcome ovarian cancer, there still remain limited optional treatments for this disease. Nobiletin, a prospective food-derived phytochemical extracted from citrus fruits, has recently been reported to suppress ovarian cancer cells, but the role of pyroptosis in ovarian carcinoma with nobiletin still remains unknown. In this study, we aim to explore the effect of nobiletin on ovarian carcinoma and further expound the underlying mechanisms of nobiletin-induced ovarian cancer cell death. Our results showed that nobiletin could significantly inhibit cell proliferation, induce DNA damage, and also lead to apoptosis by increasing the cleaved poly (ADP-ribose) polymerase (PARP) level of human ovarian cancer cells (HOCCs) in a dose-dependent manner. Moreover, we revealed that nobiletin decreased mitochondrial membrane potential and induced reactive oxygen species (ROS) generation and autophagy of HOCCs, contributing to gasdermin D-/gasdermin E-mediated pyroptosis. Taken together, nobiletin as a functional food ingredient represents a promising new anti-ovarian cancer candidate that could induce apoptosis and trigger ROS-mediated pyroptosis through regulating autophagy in ovarian cancer cells.
Collapse
Affiliation(s)
- Rongjun Zhang
- Cancer Research Institute, School of Basic Medical Sciences , Southern Medical University , Guangzhou 510515 , Guangdong , China
| | - Jian Chen
- Cancer Research Institute, School of Basic Medical Sciences , Southern Medical University , Guangzhou 510515 , Guangdong , China
| | - Lianzhi Mao
- Department of Nutrition and Food Hygiene, School of Public Health , Southern Medical University , Guangzhou 510515 , Guangdong , China
| | - Yajie Guo
- The Eighth Affiliated Hospital , Sun Yat-sen University , Shenzhen 518033 , Guangdong , China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, School of Public Health , Southern Medical University , Guangzhou 510515 , Guangdong , China
| | - Yudi Deng
- Department of Nutrition and Food Hygiene, School of Public Health , Southern Medical University , Guangzhou 510515 , Guangdong , China
| | - Xue Han
- Department of Obstetrics and Gynecology , Gansu Provincial Hospital , Lanzhou 730000 , Gansu , China
| | - Qingjiao Li
- The Eighth Affiliated Hospital , Sun Yat-sen University , Shenzhen 518033 , Guangdong , China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, School of Public Health , Southern Medical University , Guangzhou 510515 , Guangdong , China
| | - Miaomiao Yuan
- Cancer Research Institute, School of Basic Medical Sciences , Southern Medical University , Guangzhou 510515 , Guangdong , China
- The Eighth Affiliated Hospital , Sun Yat-sen University , Shenzhen 518033 , Guangdong , China
| |
Collapse
|
37
|
Zhuang J, Yin J, Xu C, Jiang M, Lv S. Diverse autophagy and apoptosis in myeloid leukemia cells induced by 20(s)-GRh2 and blue LED irradiation. RSC Adv 2019; 9:39124-39132. [PMID: 35540666 PMCID: PMC9075934 DOI: 10.1039/c9ra08049j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/12/2019] [Indexed: 11/21/2022] Open
Abstract
Autophagy is an important mechanism for cell death regulation. To improve the anticancer effect during the treatment of leukemia and promote the apoptosis of leukemic cells, it is important to define the relationship between autophagy and apoptosis. A key bioactive compound in traditional Chinese medicine, 20(s)-Ginsenoside (GRh2), demonstrated an advancement in leukemia treatment. Blue LED therapy (BL) is a physical treatment method that can induce leukemic cell death. In this study, we tested the effect of 20(s)-GRh2, BL, and their combination (BL-GRh2) on the activation of leukemic cell apoptosis and autophagy. Both treatments, whether used individually or simultaneously, induce apoptosis through the induction of reactive oxygen species (ROS), disrupted mitochondrial membrane potential (MMP) and regulated the expression of apoptosis-related genes and proteins. Furthermore, using western blotting to analyze the autophagy markers LC3B and P62, we detected the activation of autophagy. In cells treated with autophagy inhibitor 3-MA, both autophagy and apoptosis were inhibited, either by BL alone or by BL-GRh2. However, apoptosis in 20(s)-GRh2-treated cells was enhanced. In cells treated with apoptosis suppressor Z-VAD-FMK, autophagy was inhibited in the BL and BL-GRh2-treated cells, although it was enhanced in cells treated with 20(s)-GRh2 alone. Moreover, we observed a stronger induction of apoptosis by BL-GRh2 in myeloid leukemia cells. Our data indicate that autophagy induced by different factors can play diverse roles on the same cells. Our results also indicate that the combination of traditional Chinese medicine with physical therapy may be a new strategy for anti-cancer therapy.
Collapse
Affiliation(s)
- Jianjian Zhuang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University Changchun 130000 China
| | - Juxin Yin
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University Changchun 130000 China
- Research Centre for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University Hangzhou Zhejiang Province 310058 P. R. China
| | - Chaojian Xu
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University Changchun 130000 China
| | - Mengmeng Jiang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University Changchun 130000 China
| | - Shaowu Lv
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University Changchun 130000 China
| |
Collapse
|
38
|
Zheng SW, Xiao SY, Wang J, Hou W, Wang YP. Inhibitory Effects of Ginsenoside Ro on the Growth of B16F10 Melanoma via Its Metabolites. Molecules 2019; 24:E2985. [PMID: 31426477 PMCID: PMC6721120 DOI: 10.3390/molecules24162985] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/24/2022] Open
Abstract
Ginsenoside Ro (Ro), a major saponin derived and isolated from Panax ginseng C.A. Meyer, exerts multiple biological activities. However, the anti-tumour efficacy of Ro remains unclear because of its poor in vitro effects. In this study, we confirmed that Ro has no anti-tumour activity in vitro. We explored the anti-tumour activity of Ro in vivo in B16F10 tumour-bearing mice. The results revealed that Ro considerably suppressed tumour growth with no significant side effects on immune organs and body weight. Zingibroside R1, chikusetsusaponin IVa, and calenduloside E, three metabolites of Ro, were detected in the plasma of Ro-treated tumour-bearing mice and showed excellent anti-tumour effects as well as anti-angiogenic activity. The results suggest that the metabolites play important roles in the anti-tumour efficacy of Ro in vivo. Additionally, the haemolysis test demonstrated that Ro has good biocompatibility. Taken together, the findings of this study demonstrate that Ro markedly suppresses the tumour growth of B16F10-transplanted tumours in vivo, and its anti-tumour effects are based on the biological activity of its metabolites. The anti-tumour efficacy of these metabolites is due, at least in part, to its anti-angiogenic activity.
Collapse
Affiliation(s)
- Si-Wen Zheng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Sheng-Yuan Xiao
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Jia Wang
- School of Pharmaceutical Sciences Changchun University of Chinese Medicine, Changchun 130117, China
| | - Wei Hou
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Ying-Ping Wang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China.
| |
Collapse
|
39
|
Qi Z, Li W, Tan J, Wang C, Lin H, Zhou B, Liu J, Li P. Effect of ginsenoside Rh 2 on renal apoptosis in cisplatin-induced nephrotoxicity in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 61:152862. [PMID: 31048124 DOI: 10.1016/j.phymed.2019.152862] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 05/13/2023]
Abstract
BACKGROUND Ginsenoside Rh2 (Rh2), an important ingredient from Panax ginseng, has received much attention due to a range of pharmacological actions. PURPOSE The aim of the study was to investigate the therapeutic potential Rh2 on cisplatin (CDDP)-induced nephrotoxicity and to elucidate involved mechanisms. STUDY DESIGN An in vivo mice model of CDDP-induced nephrotoxicity was established by a single intraperitoneal injection of CDDP (20 mg/kg) to assess the effects of Rh2 on renal biochemical parameter, oxidative stress, inflammation tubular cell apoptosis and serum metabolic profiles. RESULTS Rh2 protected against CDDP-induced renal dysfunction and ameliorated CDDP-induced oxidative stress, histopathological damage, inflammation and tubular cell apoptosis in kidney. Rh2 treatment had significantly increased expression of Bcl-2 and decreased expression of p53, Bax, cytochrome c, caspase-8, caspase-9, and caspase-3 in kidney tissues. Metabolomic analysis identified 29 altered serum metabolites in Rh2 treatment mice. CONCLUSION These results suggest that Rh2 protects against CDDP-induced nephrotoxicity via action on caspase-mediated pathway.
Collapse
Affiliation(s)
- Zeng Qi
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jing Tan
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Cuizhu Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hongqiang Lin
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Baisong Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Jinping Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
40
|
Xia T, Zhang J, Zhou C, Li Y, Duan W, Zhang B, Wang M, Fang J. 20(S)-Ginsenoside Rh2 displays efficacy against T-cell acute lymphoblastic leukemia through the PI3K/Akt/mTOR signal pathway. J Ginseng Res 2019; 44:725-737. [PMID: 32913402 PMCID: PMC7471214 DOI: 10.1016/j.jgr.2019.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/20/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background T-cell acute lymphoblastic leukemia (T-ALL) is a kind of aggressive hematological cancer, and the PI3K/Akt/mTOR signaling pathway is activated in most patients with T-ALL and responsible for poor prognosis. 20(S)-Ginsenoside Rh2 (20(S)-GRh2) is a major active compound extracted from ginseng, which exhibits anti-cancer effects. However, the underlying anticancer mechanisms of 20(S)-GRh2 targeting the PI3K/Akt/mTOR pathway in T-ALL have not been explored. Methods Cell growth and cell cycle were determined to investigate the effect of 20(S)-GRh2 on ALL cells. PI3K/Akt/mTOR pathway–related proteins were detected in 20(S)-GRh2–treated Jurkat cells by immunoblotting. Antitumor effect of 20(S)-GRh2 against T-ALL was investigated in xenograft mice. The mechanisms of 20(S)-GRh2 against T-ALL were examined by cell proliferation, apoptosis, and autophagy. Results In the present study, the results showed that 20(S)-GRh2 decreased cell growth and arrested cell cycle at the G1 phase in ALL cells. 20(S)-GRh2 induced apoptosis through enhancing reactive oxygen species generation and upregulating apoptosis-related proteins. 20(S)-GRh2 significantly elevated the levels of pEGFP-LC3 and autophagy-related proteins in Jurkat cells. Furthermore, the PI3K/Akt/mTOR signaling pathway was effectively blocked by 20(S)-GRh2. 20(S)-GRh2 suppressed cell proliferation and promoted apoptosis and autophagy by suppressing the PI3K/Akt/mTOR pathway in Jurkat cells. Finally, 20(S)-GRh2 alleviated symptoms of leukemia and reduced the number of white blood cells and CD3 staining in the spleen of xenograft mice, indicating antitumor effects against T-ALL invivo. Conclusion These findings indicate that 20(S)-GRh2 exhibits beneficial effects against T-ALL through the PI3K/Akt/mTOR pathway and could be a natural product of novel target for T-ALL therapy.
Collapse
Affiliation(s)
- Ting Xia
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Jin Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Chuanxin Zhou
- Department of Pediatrics, The Fifth Hospital of Sun Yat Sen University, Sun Yat sen University, Zhuhai, Guangdong, China
| | - Yu Li
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Wenhui Duan
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Bo Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Min Wang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Jianpei Fang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guang Dong, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, Guang Dong, China
| |
Collapse
|
41
|
Qomaladewi NP, Kim MY, Cho JY. Autophagy and its regulation by ginseng components. J Ginseng Res 2019; 43:349-353. [PMID: 31308805 PMCID: PMC6606841 DOI: 10.1016/j.jgr.2018.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 12/25/2018] [Accepted: 12/26/2018] [Indexed: 01/01/2023] Open
Abstract
Autophagy is the sequential process whereby cell components are degraded, which can occur due to nutrient deprivation. Its regulation has an essential role in many diseases, functioning in both cell survival and cell death. Autophagy starts when mTORC1 is inhibited, resulting in the activation of several complexes to form a cargo that fuses with a lysosome, where it undergoes degradation. In this review, we describe a plant extract that is well known in Korea, namely Korean ginseng extract; we studied how its derivatives and metabolites can regulate autophagy and thus mediate the pathogenesis of certain diseases.
Collapse
Affiliation(s)
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
42
|
Liu Q, Tang X, Wang Y, Yang Y, Zhang W, Zhao Y, Zhang X. ROS changes are responsible for tributyl phosphate (TBP)-induced toxicity in the alga Phaeodactylum tricornutum. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 208:168-178. [PMID: 30677712 DOI: 10.1016/j.aquatox.2019.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 06/09/2023]
Abstract
As a newly emerging environmental contaminant, tributyl phosphate (TBP) is an additive flame retardant of high production volume that is frequently detected in biota and the environment. Despite evidence that TBP is a potential threat to marine organisms, ecotoxicology data for TBP in marine organisms at low trophic levels are scarce. In this study, the acute toxicological effect of TBP on the marine phytoplankton Phaeodactylum tricornutum was thoroughly investigated, and the possible mechanism was explored. The results showed that TBP at concentrations ≥0.2 mg L-1 significantly inhibited P. tricornutum growth in a clear dose-response manner, with 72-h EC10, EC20, EC50 and EC90 values of 0.067, 0.101, 0.219 and 0.716 mg L-1, respectively. Algal cells treated with TBP exhibited distorted shapes, ruptured cell membranes and damaged organelles, especially mitochondria. Additionally, apoptosis was triggered, followed by a decrease in mitochondrial membrane potential, indicating that cellular damage occurred during exposure. Although the activities of two antioxidant enzymes, superoxide peroxidase and catalase, were upregulated by TBP at 1.2 mg L-1, excess reactive oxygen species (ROS) and malondialdehyde still accumulated in algal cells after exposure, suggesting that the cells experienced oxidative stress. Moreover, both growth inhibition and apoptosis were positively correlated with ROS levels and were ameliorated by pretreatment with the ROS scavenger N-acetyl-l-cysteine. Taken together, the results indicate that TBP exposure leads to growth inhibition and cellular damage in P. tricornutum, and a ROS-mediated pathway might contribute to these observed toxicological effects.
Collapse
Affiliation(s)
- Qian Liu
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| | - Xuexi Tang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| | - You Wang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| | - Yingying Yang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| | - Wei Zhang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China.
| | - Yunchen Zhao
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian, China.
| | - Xinxin Zhang
- Department of Marine Ecology, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Ecology and Environmental Science, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
43
|
Qi Z, Chen L, Li Z, Shao Z, Qi Y, Gao K, Liu S, Sun Y, Li P, Liu J. Immunomodulatory Effects of (24R)-Pseudo-Ginsenoside HQ and (24S)-Pseudo-Ginsenoside HQ on Cyclophosphamide-Induced Immunosuppression and Their Anti-Tumor Effects Study. Int J Mol Sci 2019; 20:E836. [PMID: 30769948 PMCID: PMC6413033 DOI: 10.3390/ijms20040836] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
(24R)-pseudo-ginsenoside HQ (R-PHQ) and (24S)-pseudo-ginsenoside HQ (S-PHQ) are the main metabolites of (20S)-ginsenoside Rh₂ (Rh₂) in vivo. In this study, we found that Rh₂, R-PHQ, and S-PHQ upregulated the innate and adaptive immune response in cyclophosphamide (CTX) induced-immunocompromised mice as evidenced by the number of leukocytes, cellular immunity, and phagocytosis of macrophages. Spleen T-lymphocyte subpopulations and the serum cytokines level were also balanced in these immunosuppressed mice. Furthermore, co-administration with R-PHQ or S-PHQ did not compromise the antitumor activity of CTX in the hepatoma H22-bearing mice. Treatment with R-PHQ and S-PHQ clearly induced the apoptosis of tumor cells, significantly increased the expression of Bax, and remarkably inhibited the expression of Bcl-2 and vascular endothelial growth factor (VEGF) in H22 tumor tissues. The anti-tumor activity of R-PHQ and S-PHQ could be related to the promotion of tumor apoptosis and inhibition of angiogenesis and may involve the caspase and VEGF signaling pathways. This study provides a theoretical basis for further study on R-PHQ and S-PHQ.
Collapse
Affiliation(s)
- Zeng Qi
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| | - Lixue Chen
- Institute of Special Animals and Plants Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Zhuo Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| | - Zijun Shao
- Institute of Special Animals and Plants Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Yuli Qi
- Institute of Special Animals and Plants Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Kun Gao
- Institute of Special Animals and Plants Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Songxin Liu
- Institute of Special Animals and Plants Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Yinshi Sun
- Institute of Special Animals and Plants Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| | - Jinping Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
44
|
Hsieh YH, Deng JS, Chang YS, Huang GJ. Ginsenoside Rh2 Ameliorates Lipopolysaccharide-Induced Acute Lung Injury by Regulating the TLR4/PI3K/Akt/mTOR, Raf-1/MEK/ERK, and Keap1/Nrf2/HO-1 Signaling Pathways in Mice. Nutrients 2018; 10:nu10091208. [PMID: 30200495 PMCID: PMC6163254 DOI: 10.3390/nu10091208] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/16/2018] [Accepted: 08/30/2018] [Indexed: 12/19/2022] Open
Abstract
The anti-inflammatory effect of ginsenoside Rh2 (GRh2) has labeled it as one of the most important ginsenosides. The purpose of this study was to identify the anti-inflammatory and antioxidant effects of GRh2 using a lipopolysaccharide (LPS) challenge lung-injury animal model. GRh2 reduced LPS-induced proinflammatory mediator nitric oxide (NO), tumor necrosis factor-alpha, interleukin (IL)-1β, and anti-inflammatory cytokines (IL-4, IL-6, and IL-10) production in lung tissues. GRh2 treatment decreased the histological alterations in the lung tissues and bronchoalveolar lavage fluid (BALF) protein content; total cell number also reduced in LPS-induced lung injury in mice. Moreover, GRh2 blocked iNOS, COX-2, the phosphorylation of IκB-α, ERK, JNK, p38, Raf-1, and MEK protein expression, which corresponds with the growth of HO-1, Nrf-2, catalase, SOD, and GPx expression in LPS-induced lung injury. An in vivo experimental study suggested that GRh2 has anti-inflammatory effects, and has potential therapeutic efficacy in major anterior segment lung diseases.
Collapse
Affiliation(s)
- Yung-Hung Hsieh
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 413, Taiwan.
- Department of Pharmacy, Kuang Tien General Hospital, Taichung 433, Taiwan.
- Taichung City New Pharmacist Association, Taichung 420, Taiwan.
| | - Jeng-Shyan Deng
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 413, Taiwan.
| | - Yuan-Shiun Chang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 413, Taiwan.
| | - Guan-Jhong Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 413, Taiwan.
| |
Collapse
|