1
|
Zheng Y, Feng N, Li C, Li Z. Natural products target programmed cell death signaling mechanisms to treat colorectal cancer. Front Pharmacol 2025; 16:1565332. [PMID: 40342991 PMCID: PMC12058791 DOI: 10.3389/fphar.2025.1565332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
As a highly prevalent gastrointestinal malignant tumor, colorectal cancer poses a serious challenge in terms of increasing morbidity and mortality and late diagnosis due to the invisibility of the disease. Although existing therapies are diverse but limited in efficacy, the mechanism of programmed cell death (PCD) has become a focus of research due to its central role in maintaining body homeostasis and regulating tumor progression. Multimodal cell death pathways, such as apoptosis, autophagy, pyroptosis and ferroptosis, have shown unique advantages in inhibiting the proliferation and migration of colorectal cancer cells and enhancing the sensitivity to chemotherapy by responding to internal and external environmental stimuli. In recent years, natural products have risen to prominence by virtue of their multi-target synergistic effects and chemo-sensitizing properties, and have opened up a new direction for colorectal cancer treatment by precisely regulating the PCD pathway. In this paper, we searched PubMed, Web of Science and CNKI databases for relevant studies in the last 10 years using the keywords (Colorectal cancer) and (programmed cell death) and natural products. This work retrieved 59 studies (55 from the past 5 years and 4 from the past 10 years) to reveal the mechanism of action of natural products targeting PCD, aiming to provide theoretical support and practical guidance for the optimization of clinical therapeutic strategies and the development of innovative drugs.
Collapse
Affiliation(s)
- Ya Zheng
- The Second Gastroenterology Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Na Feng
- Department of Rehabilitation Medicine, Linyi Maternal and Child Health Center Hospital, Linyi, Shandong, China
| | - Canglin Li
- Medical Management Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zuoqiang Li
- Department of Traditional Chinese Medicine, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| |
Collapse
|
2
|
Jia ZH, Pilkington LI, Barker D. Total Synthesis of the Furopyran Lignans Sumatranin A-D and the Proposed Structure of Sumatranin H. JOURNAL OF NATURAL PRODUCTS 2025; 88:563-576. [PMID: 39964090 DOI: 10.1021/acs.jnatprod.4c01466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Sumatranins A-D are lignans isolated from the twigs of Cleistanthus sumatranus that contain a previously unseen furopyran in a tetrahydro-furo[2,3-b]chromene tricyclic system. In this work, sumatranins A-D were enantioselectively synthesized utilizing an Evans aldol reaction followed by acid-catalyzed cyclization as key steps. Additionally, the proposed structure of dibenzylbutyrolactone lignan sumatranin H, an apparent biosynthetic precursor to the furopyran lignans, was synthesized but determined to be inconsistent with the previously isolated data. The synthetic routes developed allows for the construction of a wide range of sumatranin-type lignans or unnatural analogues.
Collapse
Affiliation(s)
- Zong Hao Jia
- School of Chemical Sciences, University of Auckland, Auckland 1010, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Lisa I Pilkington
- School of Chemical Sciences, University of Auckland, Auckland 1010, New Zealand
- Te Pu̅naha Matatini, Auckland 1010, New Zealand
| | - David Barker
- School of Chemical Sciences, University of Auckland, Auckland 1010, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| |
Collapse
|
3
|
Wongpan A, Panvongsa W, Krobthong S, Nutho B, Kanjanasirirat P, Jearawuttanakul K, Khumpanied T, Phlaetita S, Chabang N, Munyoo B, Tuchinda P, Ponpuak M, Borwornpinyo S, Chairoungdua A. Cleistanthin A derivative disrupts autophagy and suppresses head and neck squamous cell carcinoma progression via targeted vacuolar ATPase. Sci Rep 2024; 14:22582. [PMID: 39343784 PMCID: PMC11439923 DOI: 10.1038/s41598-024-73186-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) present a significant challenge due to its heterogeneity and limited treatment options, often resulting in severe side effects and poor survival rates with conventional chemoradiotherapy. Here, we investigated the anticancer activity of halogenated benzoate derivatives of cleistanthin A, ECDD-S16 and ECDD-S18, in HNSCC cells. Our findings revealed that ECDD-S18 exhibited remarkable cytotoxicity, surpassing that of cisplatin with minimal impact on normal and cisplatin-sensitive cells. Notably, ECDD-S18 induced apoptosis in a dose-dependent manner and effectively targeted vacuolar ATPase (V-ATPase), impairing lysosomal acidification. Intriguingly, ECDD-S18 inhibited autophagic flux, as evidenced by increased autophagosome but decreased autolysosome formation. Furthermore, proteomic analysis demonstrated downregulation of cathepsin D (CTSD), the lysosomal protease in ECDD-S18-treated HNSCC cells, concurrent with suppressed cell migration. ECDD-S18 also decreased expression of mesenchymal markers, suggesting inhibition of epithelial-mesenchymal transition (EMT). Importantly, cotreatment with ECDD-S18 and cisplatin enhanced the reduction in cell viability. Collectively, our results indicated that the anticancer activity of ECDD-S18 partly stems from its ability to disrupt lysosomal acidification and inhibit autophagy via targeted inhibition of V-ATPase. These findings underscore the therapeutic promise of ECDD-S18 in HNSCC treatment, either alone or in combination with existing drugs, while mitigating toxicity to normal cells.
Collapse
Affiliation(s)
- Anongnat Wongpan
- Department of Physiology, Faculty of Science, Mahidol University, Rama 6 Rd., Ratchathewi, Bangkok, 10400, Thailand
| | - Wittaya Panvongsa
- Department of Tropical Nutrition and Food Science, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sucheewin Krobthong
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Bodee Nutho
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Phongthon Kanjanasirirat
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Tanawadee Khumpanied
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
| | - Sureeporn Phlaetita
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Napason Chabang
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Bamroong Munyoo
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
- Department of Chemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Patoomratana Tuchinda
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
- Department of Chemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Marisa Ponpuak
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Arthit Chairoungdua
- Department of Physiology, Faculty of Science, Mahidol University, Rama 6 Rd., Ratchathewi, Bangkok, 10400, Thailand.
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand.
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
4
|
Yolanda H, Jearawuttanakul K, Wannalo W, Kanjanasirirat P, Borwornpinyo S, Rujirawat T, Payattikul P, Kittichotirat W, Wichadakul D, Krajaejun T. Potential anti- Pythium insidiosum therapeutics identified through screening of agricultural fungicides. Microbiol Spectr 2024; 12:e0162023. [PMID: 38179943 PMCID: PMC10846074 DOI: 10.1128/spectrum.01620-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/07/2023] [Indexed: 01/06/2024] Open
Abstract
Pythiosis is a life-threatening infectious disease caused by the oomycete Pythium insidiosum. Clinical manifestations of pythiosis include an eye, blood vessel, skin, or gastrointestinal tract infection. Pythiosis has been increasingly reported worldwide, with an overall mortality rate of 28%. Radical surgery is required to save patients' lives due to the limited efficacy of antimicrobial drugs. Effective medical treatments are urgently needed for pythiosis. This study aims to find anti-P. insidiosum agents by screening 17 agricultural fungicides that inhibit plant-pathogenic oomycetes and validating their efficacy and safety. Cyazofamid outperformed other fungicides as it can potently inhibit genetically diverse P. insidiosum isolates while exhibiting minimal cellular toxicities. The calculated therapeutic scores determined that the concentration of cyazofamid causing significant cellular toxicities was eight times greater than the concentration of the drug effectively inhibiting P. insidiosum. Furthermore, other studies showed that cyazofamid exhibits low-to-moderate toxicities in animals. The mechanism of cyazofamid action is likely the inhibition of cytochrome b, an essential component in ATP synthesis. Molecular docking and dynamic analyses depicted a stable binding of cyazofamid to the Qi site of the P. insidiosum's cytochrome b orthologous protein. In conclusion, our search for an effective anti-P. insidiosum drug indicated that cyazofamid is a promising candidate for treating pythiosis. With its high efficacy and low toxicity, cyazofamid is a potential chemical for treating pythiosis, reducing the need for radical surgeries, and improving recovery rates. Our findings could pave the way for the development of new and effective treatments for pythiosis.IMPORTANCEPythiosis is a severe infection caused by Pythium insidiosum. The disease is prevalent in tropical/subtropical regions. This infectious condition is challenging to treat with antifungal drugs and often requires surgical removal of the infected tissue. Pythiosis can be fatal if not treated promptly. There is a need for a new treatment that effectively inhibits P. insidiosum. This study screened 17 agricultural fungicides that target plant-pathogenic oomycetes and found that cyazofamid was the most potent in inhibiting P. insidiosum. Cyazofamid showed low toxicity to mammalian cells and high affinity to the P. insidiosum's cytochrome b, which is involved in energy production. Cyazofamid could be a promising candidate for the treatment of pythiosis, as it could reduce the need for surgery and improve the survival rate of patients. This study provides valuable insights into the biology and drug susceptibility of P. insidiosum and opens new avenues for developing effective therapies for pythiosis.
Collapse
Affiliation(s)
- Hanna Yolanda
- Program in Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Department of Parasitology, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Kedchin Jearawuttanakul
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Warawuth Wannalo
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Thidarat Rujirawat
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Penpan Payattikul
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Weerayuth Kittichotirat
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bangkhuntien, Bangkok, Thailand
- Systems Biology and Bioinformatics Research Group, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bangkhuntien, Bangkok, Thailand
| | - Duangdao Wichadakul
- Department of Computer Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Theerapong Krajaejun
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
5
|
Zhou Z, Han S, Liao J, Wang R, Yu X, Li M. Isoliquiritigenin Inhibits Oral Squamous Cell Carcinoma and Overcomes Chemoresistance by Destruction of Survivin. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2023; 51:2221-2241. [PMID: 37930332 DOI: 10.1142/s0192415x23500957] [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: 11/07/2023]
Abstract
The oncoprotein survivin plays a pivotal role in controlling cell division and preventing apoptosis by inhibiting caspase activation. Its significant contribution to tumorigenesis and therapeutic resistance has been well established. Isoliquiritigenin (ISL), a natural compound, has been recognized for its powerful inhibitory effects against various tumors. However, whether ISL exerts regulatory effects on survivin and its underlying mechanism in oral squamous cell carcinoma (OSCC) remains unclear. Here, we found that ISL inhibited the viability and colony formation of OSCC, and promoted their apoptosis. The immunoblotting data showed that ISL treatment significantly decreased survivin expression. Mechanistically, ISL suppressed survivin phosphorylation on Thr34 by deregulating Akt-Wee1-CDK1 signaling, which facilitated survivin for ubiquitination degradation. ISL inhibited CAL27 tumor growth and decreased p-Akt and survivin expression in vivo. Meanwhile, survivin overexpression caused cisplatin resistance of OSCC cells. ISL alone or combined with cisplatin overcame chemoresistance in OSCC cells. Overall, our results revealed that ISL exerted potent inhibitory effects via inducing Akt-dependent survivin ubiquitination in OSCC cells.
Collapse
Affiliation(s)
- Zhongsu Zhou
- The Third Hospital of Changsha, Changsha, Hunan 410015, P. R. China
| | - Shuangze Han
- The Third Hospital of Changsha, Changsha, Hunan 410015, P. R. China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P. R. China
| | - Jinzhuang Liao
- The Third Hospital of Changsha, Changsha, Hunan 410015, P. R. China
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P. R. China
| | - Ruirui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P. R. China
| | - Xinfang Yu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ming Li
- Hunan University of Chinese Medicine, Affiliated Stomatological Hospital, Changsha, Hunan 410208, P. R. China
- Changsha Stomatological Hospital, Changsha, Hunan 410004, P. R. China
| |
Collapse
|
6
|
Bay MV, Nam PC, Hoa NT, Mechler A, Vo QV. Antiradical Activity of Lignans from Cleistanthus sumatranus: Theoretical Insights into the Mechanism, Kinetics, and Solvent Effects. ACS OMEGA 2023; 8:38668-38675. [PMID: 37867707 PMCID: PMC10586290 DOI: 10.1021/acsomega.3c05964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023]
Abstract
Sumatranus lignans (SL) isolated from Cleistanthus sumatranus have demonstrated bioactivities, e.g., they were shown to exhibit immunosuppressive properties in previous research. Their structure suggests potential antioxidant activity that has not attracted any attention thus far. Consistently, a comprehensive analysis of the antioxidant activity of these compounds is highly desirable with the view of prospective medical applications. In this work, the mechanism and kinetics of the antiradical properties of SL against hydroperoxyl radicals were studied by using calculations based on density functional theory (DFT). In the lipid medium, it was discovered that SL reacted with HOO• through the formal hydrogen transfer mechanism with a rate constant of 101-105 M-1 s-1, whereas in aqueous media, the activity primarily occurred through the sequential proton loss electron transfer mechanism with rate constants of 102-108 M-1 s-1. In both lipidic and aqueous environments, the antiradical activity of compounds 6 and 7 exceeds that of resveratrol, ascorbic acid, and Trolox. These substances are therefore predicted to be good radical scavengers in physiological environments.
Collapse
Affiliation(s)
- Mai Van Bay
- The
University of Danang - University of Science and Education, Danang 550000, Vietnam
| | - Pham Cam Nam
- The
University of Danang - University of Science and Technology, Danang 550000, Vietnam
| | - Nguyen Thi Hoa
- The
University of Danang - University of Technology and Education, Danang 550000, Vietnam
| | - Adam Mechler
- Department
of Biochemistry and Chemistry, La Trobe
University, Victoria 3086, Australia
| | - Quan V. Vo
- The
University of Danang - University of Technology and Education, Danang 550000, Vietnam
| |
Collapse
|
7
|
Zeng JN, Tan JY, Mo L. Naringin Inhibits Colorectal Carcinogenesis by Inhibiting Viability of Colorectal Cancer Cells. Chin J Integr Med 2023; 29:707-713. [PMID: 37340202 DOI: 10.1007/s11655-023-3698-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2023] [Indexed: 06/22/2023]
Abstract
OBJECTIVE To explore the therapeutic effect of naringin on colorectal cancer (CRC) and the related mechanism. METHODS Cell counting kit-8 (CCK-8) assay and annexin V-FITC/PI assay were used to detect the effect of naringin (50-400 µg/mL) on cell proliferation and apoptosis of CRC cells, respectively. The scratch wound assay and transwell migration assay were used to assess the effect of naringin on CRC cell migration. Four-week-old male nude mice were injected with HCT116 cells subcutaneously to establish the tumor xenograft model. Naringin was injected intraperitoneally at 50 mg/(kg·d), with solvent and 5-fluorouracil treatment as control. The width and length of the tumors were measured and recorded every 6 days, and tumor tissues were photographed and weighed on the last day of the 24-d observation period. Immunohistochemical staining for caspase-3, proliferating cell nuclear antigen and TUNEL assay were used to evaluate the effect of naringin on cell proliferation and apoptosis in tumor tissues. The body weight, food and water intake of mice were recorded, and the major organs in different treatment groups were weighed on the last day and stained with hematoxylin and eosin for histological analysis. Meanwhile, the routine blood indicators were recorded. RESULTS CCK-8 and annexin V-FITC/PI results confirmed that naringin (100, 200, and 400 µg/mL) could inhibit proliferation and promote apoptosis. The scratch wound assay and transwell migration assay results confirmed the inhibitory activity of naringin against CRC cells migration. In vivo results demonstrated the inhibitory effect of naringin on tumor growth with good bio-compatibility. CONCLUSION Naringin inhibited colorectal carcinogenesis by inhibiting viability of CRC cells.
Collapse
Affiliation(s)
- Juan-Ni Zeng
- Department of Anus-Intestines, the Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410008, China
| | - Jin-Yu Tan
- Department of Anus-Intestines, the Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410008, China
| | - Li Mo
- Department of Anus-Intestines, the Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410008, China.
| |
Collapse
|
8
|
Zhou JS, Yao JY, Gao Y, Liu QF, Zhou B, He SJ, Zhao JX, Yue JM. Sumatranins A-J: Lignans with Immunosuppressive Activity from Cleistanthus sumatranus. JOURNAL OF NATURAL PRODUCTS 2023; 86:1606-1614. [PMID: 37307145 DOI: 10.1021/acs.jnatprod.3c00300] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Chemical investigation of the twigs of Cleistanthus sumatranus (Phyllanthaceae) led to the isolation of 10 undescribed lignans, sumatranins A-J (1-10). Compounds 1-4 are unprecedented furopyran lignans characterized by a unique 2,3,3a,9a-tetrahydro-4H-furo[2,3-b]chromene heterotricyclic framework. Compounds 9 and 10 are rare 9'-nor-dibenzylbutane lignans. Structures were established based on analyses of spectroscopic data, X-ray crystallographic data, and experimental ECD spectra. Immunosuppressive assays revealed compounds 3 and 9 displayed moderate inhibitory effects with good selectivity indexes against LPS-induced B lymphocyte proliferation.
Collapse
Affiliation(s)
- Jun-Su Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, People's Republic of China
| | - Jia-Ying Yao
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, 818 Meiling Avenue, Nanchang, Jiangxi 330004, People's Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| | - Yuan Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, People's Republic of China
| | - Qun-Fang Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, People's Republic of China
| | - Bin Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, People's Republic of China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, 198 East Binhai Road, Yantai, Shandong 264117, People's Republic of China
| | - Shi-Jun He
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| | - Jin-Xin Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, People's Republic of China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, 198 East Binhai Road, Yantai, Shandong 264117, People's Republic of China
| | - Jian-Min Yue
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, People's Republic of China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, 198 East Binhai Road, Yantai, Shandong 264117, People's Republic of China
| |
Collapse
|
9
|
Lin X, Chen J, Li X, Chen D, Luo K, Deng Y, Yang D, Huang Z, Tao C. Dimeric oxyberberine CT4-1 targets LINC02331 to induce cytotoxicity and inhibit chemoresistance via suppressing Wnt/β-catenin signaling in hepatocellular carcinoma. Arch Toxicol 2023; 97:1627-1647. [PMID: 37120773 DOI: 10.1007/s00204-023-03501-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/20/2023] [Indexed: 05/01/2023]
Abstract
Hepatocellular carcinoma (HCC) is a type of cancer characterized by high recurrence rates. Overcoming chemoresistance can reduce HCC recurrence and improve patients' prognosis. This work aimed to identify HCC chemoresistance-associated long non-coding RNA (lncRNA) and find an effective drug targeting the identified lncRNA for ameliorating the chemoresistance. In this investigation, bioinformatics analysis based on The Cancer Genome Atlas revealed a new chemoresistance index and suggested LINC02331 as an HCC chemoresistance and patients' prognosis-associated lncRNA that served as an independent prognostic indicator. Moreover, LINC02331 promoted DNA damage repair, DNA replication, and epithelial-mesenchymal transition as well as attenuated cell cycle arrest and apoptosis through regulating Wnt/β-catenin signaling, thus stimulating HCC resistance to cisplatin cytotoxicity, proliferation, and metastasis. Interestingly, we developed a novel oxidative coupling approach to synthesize a dimeric oxyberberine CT4-1, which exerted superior anti-HCC activities without obvious side effects measured by in vivo mice model and could downregulate LINC02331 mice model and could downregulate LINC02331 to mitigate LINC02331-induced HCC progression by suppressing Wnt/β-catenin signaling. RNA sequencing analyses verified the involvement of CT4-1-affected differential expression genes in dysregulated pathways and processes, including Wnt, DNA damage repair, cell cycle, DNA replication, apoptosis, and cell adhesion molecules. Furthermore, CT4-1 was demonstrated to be an effective cytotoxic drug in ameliorating HCC patients' prognosis with a prediction model constructed based on RNA-sequencing data from CT4-1-treated cancer cells and public cancer database. In summary, HCC chemoresistance-associated LINC02331 independently predicted poor patients' prognosis and enhanced HCC progression by promoting resistance to cisplatin cytotoxicity, proliferation, and metastasis. Targeting LINC02331 by the dimeric oxyberberine CT4-1 that exhibited synergistic cytotoxicity with cisplatin could alleviate HCC progression and improve patients' prognosis. Our study identified LINC02331 as an alternative target and suggested CT4-1 as an effective cytotoxic drug in HCC treatment.
Collapse
Affiliation(s)
- Xian Lin
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Jian Chen
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, 518036, China
- Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Xin Li
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Dong Chen
- Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Kaixuan Luo
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Yongxing Deng
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Dinghua Yang
- Unit of Hepatobiliary Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Zunnan Huang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan, 523808, China.
| | - Cheng Tao
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
10
|
Liu C, Wang S, Xiang Z, Xu T, He M, Xue Q, Song H, Gao P, Cong Z. The chemistry and efficacy benefits of polysaccharides from Atractylodes macrocephala Koidz. Front Pharmacol 2022; 13:952061. [PMID: 36091757 PMCID: PMC9452894 DOI: 10.3389/fphar.2022.952061] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
Atractylodes macrocephala Koidz (AM), traditional Chinese medicine (TCM) with many medicinal values, has a long usage history in China and other oriental countries. The phytochemical investigation revealed the presence of volatile oils, polysaccharides, lactones, flavonoids, and others. The polysaccharides from AM are important medicinal components, mainly composed of glucose (Glc), galactose (Gal), rhamnose (Rha), arabinose (Ara), mannose (Man), galacturonic acid (GalA) and xylose (Xyl). It also showed valuable bioactivities, such as immunomodulatory, antitumour, gastroprotective and intestinal health-promoting, hepatoprotective, hypoglycaemic as well as other activities. At the same time, based on its special structure and pharmacological activity, it can also be used as immune adjuvant, natural plant supplement and vaccine adjuvant. The aim of this review is to summarize and critically analyze up-to-data on the chemical compositions, biological activities and applications of polysaccharide from AM based on scientific literatures in recent years.
Collapse
Affiliation(s)
- Congying Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shengguang Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zedong Xiang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tong Xu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengyuan He
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qing Xue
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huaying Song
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peng Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Peng Gao, ; Zhufeng Cong,
| | - Zhufeng Cong
- Shandong First Medical University Affiliated Shandong Tumor Hospital and Institute, Shandong Cancer Hospital and Institute, Jinan, China
- *Correspondence: Peng Gao, ; Zhufeng Cong,
| |
Collapse
|