1
|
Zhang W, Liu Z, Hao P, Zhang P, Pei X, Shi M, Zhu Z. Polydatin protects against leukemia by inducing apoptosis and activating autophagy via p38-MAPK pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04160-5. [PMID: 40229602 DOI: 10.1007/s00210-025-04160-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
The clinical treatment of acute myeloid leukemia (AML) remains suboptimal, necessitating the exploration of novel therapeutic strategies. Polydatin, a major active component of the Chinese herb Polygonum cuspidatum, exhibits multiple antitumor properties. However, its potential anti-AML effects and underlying mechanisms remain unclear. In this study, MOLM-13 cells, a representative cell line for AML, were treated with polydatin, and its effects on cell proliferation, cell cycle, apoptosis, reactive oxygen species (ROS) generation, and mitochondrial membrane potential (Δψm) were assessed using the Cell Counting Kit-8 (CCK-8) assay and flow cytometry. The expression levels of key apoptotic and autophagic markers, including death receptors (DR4 and DR5), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), Bcl-2, Bax, LC3, Beclin1, and P62, were analyzed by western blotting. Furthermore, autophagy levels were evaluated using GFP-LC3 fluorescence and monodansylcadaverine staining. To elucidate the relationship between polydatin and autophagy, the autophagy inhibitor 3-MA and the p38-MAPK activator SB203580 were employed. Polydatin significantly inhibited AML cell proliferation, induced apoptosis and autophagic flux, caused cell cycle arrest in the S phase, reduced Δψm, and promoted ROS generation. Following polydatin treatment, the protein expression levels of DR4, TRAIL, Bax, LC3, and Beclin1 were significantly increased, while DR5, Bcl-2, and P62 were markedly reduced. Additionally, SB203580 promoted polydatin's effects on cell proliferation inhibition, ROS generation, and autophagic flux, whereas 3-MA reversed these effects. These findings demonstrate that polydatin exerts anti-leukemic effects by inhibiting cell proliferation and inducing apoptosis through the activation of autophagy via the p38-MAPK pathway in MOLM-13 AML cells. This suggests that polydatin could serve as a potential therapeutic agent for AML.
Collapse
Grants
- 82570184 National Natural Science Foundation of P.R. China
- 82570184 National Natural Science Foundation of P.R. China
- 82570184 National Natural Science Foundation of P.R. China
- 82570184 National Natural Science Foundation of P.R. China
- 82570184 National Natural Science Foundation of P.R. China
- 82570184 National Natural Science Foundation of P.R. China
- 82570184 National Natural Science Foundation of P.R. China
- JQRC2023014, LHGJ20230016, SBGJ202102049, SBGJ202102041, 242102311116 Health Bureau of Henan Province, P.R. China
- JQRC2023014, LHGJ20230016, SBGJ202102049, SBGJ202102041, 242102311116 Health Bureau of Henan Province, P.R. China
- JQRC2023014, LHGJ20230016, SBGJ202102049, SBGJ202102041, 242102311116 Health Bureau of Henan Province, P.R. China
- JQRC2023014, LHGJ20230016, SBGJ202102049, SBGJ202102041, 242102311116 Health Bureau of Henan Province, P.R. China
- JQRC2023014, LHGJ20230016, SBGJ202102049, SBGJ202102041, 242102311116 Health Bureau of Henan Province, P.R. China
- JQRC2023014, LHGJ20230016, SBGJ202102049, SBGJ202102041, 242102311116 Health Bureau of Henan Province, P.R. China
- JQRC2023014, LHGJ20230016, SBGJ202102049, SBGJ202102041, 242102311116 Health Bureau of Henan Province, P.R. China
Collapse
Affiliation(s)
- Wenhui Zhang
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450002, Henan, China
| | - Zhongwen Liu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450002, Henan, China
| | - Peiyuan Hao
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450002, Henan, China
| | - Ping Zhang
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450002, Henan, China
| | - Xiaohang Pei
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450002, Henan, China
| | - Mingyue Shi
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450002, Henan, China.
| | - Zunmin Zhu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, 450002, Henan, China.
| |
Collapse
|
2
|
Faraji-Barhagh A, Jahandar-Lashaki S, Esfahlan RJ, Alizadeh E. Current nano drug delivery systems for targeting head and neck squamous cell carcinoma microenvironment: a narrative review. Mol Biol Rep 2025; 52:369. [PMID: 40195238 DOI: 10.1007/s11033-025-10462-x] [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: 02/09/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
The treatment of head and neck squamous cell carcinoma (HNSCC) remains a significant hurdle in clinical oncology, primarily due to the tumor's intricate and immune-suppressing environment, its diverse genetic and observable characteristics, and its tendency to spread locally and to distant sites, further complicated by the development of drug insensitivity. Standard treatment approaches frequently fall short in effectively managing these complex features. This article provides a critical assessment of the developing area of sophisticated drug delivery methods (DDSs) aimed at improving treatment results in HNSCC. The specific attributes of the HNSCC tumor environment are examined, with a focus on the disrupted structure of the extracellular matrix (ECM), its involvement in the spread of tumor cells through the bloodstream and the establishment of metastatic tumors, and the various ways in which drug resistance arises. Additionally, we assess how novel DDS technologies might overcome these challenges through directed delivery to particular tumor microenvironment targets, precise control of cancer-driving signaling pathways, and the avoidance of drug resistance mechanisms. This overview summarizes recent progress in DDS technologies customized for HNSCC treatment, with a particular emphasis on therapies using nanoparticles and immune-based drug delivery, highlighting their potential to address the many difficulties associated with this difficult-to-treat cancer. We will explore the progression of these treatment strategies from laboratory research to clinical practice and the ongoing efforts to improve patient survival.
Collapse
Affiliation(s)
- Aref Faraji-Barhagh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Rana Jahanban Esfahlan
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Effat Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [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: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
4
|
Donia T, Ali EMM, Kalantan AA, Alzahrani FA, Eid TM, Khamis AA. Synergistic anticancer efficacy of polydatin and sorafenib against the MCF-7 breast cancer cell line via inhibiting of PI3K/AKT/mTOR pathway and reducing resistance to treatment. Biochem Biophys Res Commun 2024; 739:150972. [PMID: 39541924 DOI: 10.1016/j.bbrc.2024.150972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Polydatin (PD), a glucoside derivative of resveratrol, has been investigated for its potential to mitigate sorafenib (SOF) side effects and combat multidrug resistance in cancer treatment. The study evaluated its mechanism of action for inhibiting the protein kinase B/mTOR pathway in promoting breast cancer proliferation. The combined PD and SOF have synergistic effects with a combination index (CI) < 1 in the liver (HepG2) and breast (MCF-7) cancer cell lines. Molecular docking studies were conducted to analyze interactions of PD& SOF with protein kinases as well as apoptotic and multidrug resistance proteins, including AKT1, PI3K, mTOR, Apaf-1, and ABCB1 in MCF-7 cells. Experimental validation through real-time PCR confirmed. PD has a strong binding affinity, particularly with AKT1 (-56 kcal/mol) and ABCB1 (-27.16 kcal/mol), a gene associated with multidrug resistance. These interactions were linked to anti-proliferative anti-angiogenic effects and reduced resistance to treatment, demonstrating PD has potential therapeutic benefits. Furthermore, PD combined with SOF induced apoptosis, inhibited cell growth, and arrested MCF-7 cells in the sub-G1 phase with increased intracellular ROS. This was accompanied by reduced expression of AKT1 and ABCB1 genes, reinforcing the anticancer efficacy of PD/SOF combination therapy. In conclusion, the findings suggest that PD/SOF could serve as a promising anticancer treatment strategy, warranting further investigation for potential clinical applications and mechanistic studies in vivo.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Ehab M M Ali
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt; Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Abdulaziz A Kalantan
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Faisal Ay Alzahrani
- Department of Chemistry, College of Sciences & Arts, King Abdulaziz University, 21911 Rabigh, Saudi Arabia.
| | - Thamir M Eid
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Abeer A Khamis
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
5
|
Ren QL, Li XL, Tian T, Li S, Shi RY, Wang Q, Zhu Y, Wang M, Hu H, Liu JG. Application of Natural Medicinal Plants Active Ingredients in Oral Squamous Cell Carcinoma. Chin J Integr Med 2024; 30:852-864. [PMID: 38607612 DOI: 10.1007/s11655-024-3804-7] [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: 12/18/2023] [Indexed: 04/13/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignant cancer of the head and neck, with high morbidity and mortality, ranking as the sixth most common cancer in the world. The treatment of OSCC is mainly radiotherapy, chemotherapy and surgery, however, the prognosis of patients is still poor and the recurrence rate is high. This paper reviews the range of effects of natural medicinal plant active ingredients (NMPAIs) on OSCC cancer, including the types of NMPAIs, anti-cancer mechanisms, involved signaling pathways, and clinical trials. The NMPAIs include terpenoids, phenols, flavonoids, glycosides, alkaloids, coumarins, and volatile oils. These active ingredients inhibit proliferation, induce apoptosis and autophagy, inhibit migration and invasion of OSCC cells, and regulate cancer immunity to exert anti-cancer effects. The mechanism involves signaling pathways such as mitogen-activated protein kinase, phosphatidylinositol 3 kinase/protein kinase B, nuclear factor kappa B, miR-22/WNT1/β-catenin and Nrf2/Keap1. Clinically, NMPAIs can inhibit the growth of OSCC, and the combined drug is more effective. Natural medicinal plants are promising candidates for the treatment of OSCC.
Collapse
Affiliation(s)
- Qun-Li Ren
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Xiao-Lan Li
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Tian Tian
- School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Shuang Li
- School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Rong-Yi Shi
- School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Qian Wang
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Yuan Zhu
- School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Miao Wang
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Huan Hu
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Jian-Guo Liu
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China.
| |
Collapse
|
6
|
Wu S, Zhao Q, Liu S, Kuang J, Zhang J, Onga A, Shen Y, Wang J, Sui H, Ni L, Ye Y, Tu X, Le HB, Zheng Y, Cui R, Zhu W. Polydatin, a potential NOX5 agonist, synergistically enhances antitumor activity of cisplatin by stimulating oxidative stress in non‑small cell lung cancer. Int J Oncol 2024; 65:77. [PMID: 38873997 PMCID: PMC11251743 DOI: 10.3892/ijo.2024.5665] [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: 11/09/2023] [Accepted: 05/10/2024] [Indexed: 06/15/2024] Open
Abstract
Non‑small cell lung cancer (NSCLC) is one of the major causes of cancer‑related death worldwide. Cisplatin is a front‑line chemotherapeutic agent in NSCLC. Nevertheless, subsequent harsh side effects and drug resistance limit its further clinical application. Polydatin (PD) induces apoptosis in various cancer cells by generating reactive oxygen species (ROS). However, underlying molecular mechanisms of PD and its effects on cisplatin‑mediated antitumor activity in NSCLC remains unknown. MTT, colony formation, wound healing analyses and flow cytometry was employed to investigate the cell phenotypic changes and ROS generation. Relative gene and protein expressions were evaluated by reverse transcription‑quantitative PCR and western blot analyses. The antitumor effects of PD, cisplatin and their combination were evaluated by mouse xenograft model. In the present study, it was found that PD in combination with cisplatin synergistically enhances the antitumor activity in NSCLC by stimulating ROS‑mediated endoplasmic reticulum stress, and the C‑Jun‑amino‑terminal kinase and p38 mitogen‑activated protein kinase signaling pathways. PD treatment elevated ROS generation by promoting expression of NADPH oxidase 5 (NOX5), and NOX5 knockdown attenuated ROS‑mediated cytotoxicity of PD in NSCLC cells. Mice xenograft model further confirmed the synergistic antitumor efficacy of combined therapy with PD and cisplatin. The present study exhibited a superior therapeutic strategy for some patients with NSCLC by combining PD and cisplatin.
Collapse
Affiliation(s)
- Siyuan Wu
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Qi Zhao
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Shengjuan Liu
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jiayang Kuang
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Ji Zhang
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Annabeth Onga
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yiwei Shen
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Jiaying Wang
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Hehuan Sui
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Lianli Ni
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yuxin Ye
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Xinyue Tu
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Han-Bo Le
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
| | - Yihu Zheng
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Ri Cui
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Wangyu Zhu
- Cellular and Molecular Biology Laboratory, Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316020, P.R. China
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| |
Collapse
|
7
|
Sheng X, Li X, Qian Y, Wang S, Xiao C. ETS1 regulates NDRG1 to promote the proliferation, migration, and invasion of OSCC. Oral Dis 2024; 30:977-990. [PMID: 36718855 DOI: 10.1111/odi.14527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 12/31/2022] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the molecular mechanism by which the transcription factor ETS1 regulates N-myc downstream regulatory gene 1 (NDRG1) to provide a new theoretical basis for the study of oral squamous cell carcinoma (OSCC). METHODS In this study, eight human OSCC and paraneoplastic samples were collected. The expressions of NDRG1, ETS1, and Ki67 were detected by immunohistochemistry; apoptosis was detected by tdt-mediated dUTP notched end labeling; cell migration and invasion were detected by Transwell; quantitative real-time PCR was performed to detect the expression of NDRG1; RNA-binding protein immunoprecipitation (RIP) assays detected NDRG1 expression; immunofluorescence assays detected ETS1 expression. RESULTS NDRG1 and ETS1 expression was significantly upregulated in cancer tissues and CAL-27 and SCC-6 cells. Knockdown of NDRG1 and ETS1 inhibited cell proliferation, migration, invasion, cloning, and EMT while promoting apoptosis and inhibited tumor development; ETS1 positively regulated NDRG1 expression; Finally, overexpression of NDRG1 in vivo and in vitro reversed the effect of ETS1 knockdown on CAL-27 and SCC-6 cells. CONCLUSIONS ETS1 positively regulates the expression of NDRG1 and promotes OSCC. Therefore, ETS1 may serve as a new target for the clinical diagnosis and treatment of OSCC.
Collapse
Affiliation(s)
- Xun Sheng
- School of Medicine, Yunnan University, Kunming, China
| | - Xudong Li
- Department of Prosthodontics of Kunming Medical University, Stomatology Hospital of Kunming Medical University, Kunming, China
| | - Yemei Qian
- Department of Oral and Maxillofacial Surgery of Kunming Medical University, Stomatology Hospital of Kunming Medical University, Kunming, China
| | - Shuhui Wang
- Department of General Dentistry of Kunming Medical University, Stomatology Hospital of Kunming Medical University, Kunming, China
| | - Chunjie Xiao
- School of Medicine, Yunnan University, Kunming, China
| |
Collapse
|
8
|
Fakhri S, Moradi SZ, Faraji F, Kooshki L, Webber K, Bishayee A. Modulation of hypoxia-inducible factor-1 signaling pathways in cancer angiogenesis, invasion, and metastasis by natural compounds: a comprehensive and critical review. Cancer Metastasis Rev 2024; 43:501-574. [PMID: 37792223 DOI: 10.1007/s10555-023-10136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Tumor cells employ multiple signaling mediators to escape the hypoxic condition and trigger angiogenesis and metastasis. As a critical orchestrate of tumorigenic conditions, hypoxia-inducible factor-1 (HIF-1) is responsible for stimulating several target genes and dysregulated pathways in tumor invasion and migration. Therefore, targeting HIF-1 pathway and cross-talked mediators seems to be a novel strategy in cancer prevention and treatment. In recent decades, tremendous efforts have been made to develop multi-targeted therapies to modulate several dysregulated pathways in cancer angiogenesis, invasion, and metastasis. In this line, natural compounds have shown a bright future in combating angiogenic and metastatic conditions. Among the natural secondary metabolites, we have evaluated the critical potential of phenolic compounds, terpenes/terpenoids, alkaloids, sulfur compounds, marine- and microbe-derived agents in the attenuation of HIF-1, and interconnected pathways in fighting tumor-associated angiogenesis and invasion. This is the first comprehensive review on natural constituents as potential regulators of HIF-1 and interconnected pathways against cancer angiogenesis and metastasis. This review aims to reshape the previous strategies in cancer prevention and treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6714415153, Iran
| | - Kassidy Webber
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA.
| |
Collapse
|
9
|
Li Y, Yu Y, Zhang L, Li Y, Gou M. Integrated transcriptomic and metabolomic analyses reveal the toxic effects of dimethoate on green vegetable soya bean seedlings. Gene 2024; 891:147799. [PMID: 37739194 DOI: 10.1016/j.gene.2023.147799] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023]
Abstract
The insecticide dimethoate, an organophosphate, has been used on crops, soybeans, fruits, and vegetables since the 1960s and is considered one of the most widely used pesticides. However, the understanding of the molecular mechanisms of dimethoate in crops, especially crop seedlings, is still limited. The green vegetable soya bean (Glycine max merr) is usually used as a vegetable-like fruit of soybean in many Asian countries. This study aimed to analyze the effect of dimethoate on the growth of green vegetable soya bean seedlings at the metabolic and transcriptional levels. An integrated analysis of the transcriptome and metabolome was performed to determine the responses of green vegetable soya bean seedlings to different concentrations (D1 for low dose, D2 for high dose and C for control) of dimethoate. In omics analyses, 4156 differentially expressed genes (DEGs) and 1935 differentially abundant metabolites (DAMs) were identified in the D1/C comparison, and 11,162 DEGs and 819 DAMs were identified in D2/C. Correlation analyses revealed dimethoate affected the metabolic pathways of green vegetable soya beans such as the biosynthesis of secondary metabolites and microbial metabolism in diverse environmental pathways, demonstrating that even small doses of dimethoate can affect green vegetable soya bean seedlings in a short period of time. Our study further enriches our understanding of the molecular mechanisms by which green vegetable soya beans are treated with dimethoate and provides a deeper understanding of the effects of dimethoate on crops.
Collapse
Affiliation(s)
- Yitong Li
- College of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Yang Yu
- College of Life Science, Liaoning Normal University, Dalian 116081, China; Department of Urology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116001, China
| | - Linqing Zhang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Yongfeng Li
- Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Meng Gou
- College of Life Science, Liaoning Normal University, Dalian 116081, China.
| |
Collapse
|
10
|
Yi Y, Zhou B, Man T, Xu Z, Tang H, Li J, Sun Z. Resveratrol Inhibits Nasopharyngeal Carcinoma (NPC) by Targeting the MAPK Signaling Pathway. Anticancer Agents Med Chem 2024; 24:1207-1219. [PMID: 38988166 DOI: 10.2174/0118715206319761240705115109] [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: 04/19/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND With conventional cancer treatments facing limitations, interest in plant-derived natural products as potential alternatives is increasing. Although resveratrol has demonstrated antitumor effects in various cancers, its impact and mechanism on nasopharyngeal carcinoma remain unclear. OBJECTIVE This study aimed to systematically investigate the anti-cancer effects of resveratrol on nasopharyngeal carcinoma using a combination of experimental pharmacology, network pharmacology, and molecular docking approaches. METHODS CCK-8, scratch wound, and transwell assays were employed to confirm the inhibitory effect of resveratrol on the proliferation, migration, and invasion of nasopharyngeal carcinoma cells. H&E and TUNEL stainings were used to observe the morphological changes and apoptosis status of resveratrol-treated cells. The underlying mechanisms were elucidated using a network pharmacology approach. Immunohistochemistry and Western blotting were utilized to validate key signaling pathways. RESULTS Resveratrol inhibited the proliferation, invasion, and migration of nasopharyngeal carcinoma cells, ultimately inducing apoptosis in a time- and dose-dependent manner. Network pharmacology analysis revealed that resveratrol may exert its anti-nasopharyngeal carcinoma effect mainly through the MAPK pathway. Immunohistochemistry results from clinical cases showed MAPK signaling activation in nasopharyngeal carcinoma tissues compared to adjacent tissues. Western blotting validated the targeting effect of resveratrol, demonstrating significant inhibition of the MAPK signaling pathway. Furthermore, molecular docking supported its multi-target role with MAPK, TP53, PIK3CA, SRC, etc. Conclusion: Resveratrol has shown promising potential in inhibiting human nasopharyngeal carcinoma cells by primarily targeting the MAPK pathway. These findings position resveratrol as a potential therapeutic agent for nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Yujuan Yi
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| | - Bo Zhou
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| | - Tengjun Man
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| | - Zihan Xu
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| | - Hong Tang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jia Li
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zheng Sun
- Institute (College) of Integrated Medicine, Dalian Medical University, Dalian, P.R. China
| |
Collapse
|
11
|
Angellotti G, Di Prima G, Belfiore E, Campisi G, De Caro V. Chemopreventive and Anticancer Role of Resveratrol against Oral Squamous Cell Carcinoma. Pharmaceutics 2023; 15:275. [PMID: 36678905 PMCID: PMC9866019 DOI: 10.3390/pharmaceutics15010275] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most prevailing and aggressive head and neck cancers, featuring high morbidity and mortality. The available conventional treatments suffer from several adverse effects and are often inefficient in terms of their survival rates. Thus, seeking novel therapeutic agents and adjuvants is of the utmost importance for modern society. Natural polyphenolic compounds have recently emerged as promising chemopreventive and anticancer agents. Specifically, the natural compound resveratrol (RSV) has recently gained momentum for this purpose. RSV is useful for treating OSCC due to its antiproliferative, antimetastatic, and proapoptotic effects. Additionally, RSV acts against tumor cells while synergically cooperating with chemotherapeutics, overcoming drug resistance phenomena. Despite these wide-spectrum effects, there are few specific investigations regarding RSV's effects against OSCC animal models that consider different routes and vehicles for the administration of RSV. Interestingly, an injectable RSV-loaded liposome-based formulation was proven to be effective against both in vitro and in vivo OSCC models, demonstrating that the development of RSV-loaded drug delivery systems for systemic and/or loco-regional applications may be the turning point in oral cancer treatment, leading to benefits from both RSV's properties as well as from targeted delivery. Given these premises, this review offers a comprehensive overview of the in vitro and in vivo effects of RSV and its main derivative, polydatin (PD), against OSCC-related cell lines and animal models, aiming to guide the scientific community in regard to RSV and PD use in the treatment of oral precancerous and cancerous lesions.
Collapse
Affiliation(s)
- Giuseppe Angellotti
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via L. Giuffrè 5, 90127 Palermo, Italy
| | - Giulia Di Prima
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Elena Belfiore
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via L. Giuffrè 5, 90127 Palermo, Italy
| | - Giuseppina Campisi
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via L. Giuffrè 5, 90127 Palermo, Italy
| | - Viviana De Caro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
12
|
Karami A, Fakhri S, Kooshki L, Khan H. Polydatin: Pharmacological Mechanisms, Therapeutic Targets, Biological Activities, and Health Benefits. Molecules 2022; 27:6474. [PMID: 36235012 PMCID: PMC9572446 DOI: 10.3390/molecules27196474] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
Polydatin is a natural potent stilbenoid polyphenol and a resveratrol derivative with improved bioavailability. Polydatin possesses potential biological activities predominantly through the modulation of pivotal signaling pathways involved in inflammation, oxidative stress, and apoptosis. Various imperative biological activities have been suggested for polydatin towards promising therapeutic effects, including anticancer, cardioprotective, anti-diabetic, gastroprotective, hepatoprotective, neuroprotective, anti-microbial, as well as health-promoting roles on the renal system, the respiratory system, rheumatoid diseases, the skeletal system, and women's health. In the present study, the therapeutic targets, biological activities, pharmacological mechanisms, and health benefits of polydatin are reviewed to provide new insights to researchers. The need to develop further clinical trials and novel delivery systems of polydatin is also considered to reveal new insights to researchers.
Collapse
Affiliation(s)
- Ahmad Karami
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Leila Kooshki
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
13
|
Polydatin Attenuates Cisplatin-Induced Acute Kidney Injury via SIRT6-Mediated Autophagy Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9035547. [PMID: 36160707 PMCID: PMC9507782 DOI: 10.1155/2022/9035547] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/17/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022]
Abstract
In the treatment of malignant tumors, the effectiveness of cisplatin (CP) is limited by its nephrotoxicity, leading to cisplatin-induced acute kidney injury (CP-AKI). Polydatin (PD) has been demonstrated to regulate autophagy in tumors, sepsis, and diabetes. We have recently confirmed that PD attenuated CP-AKI by inhibiting ferroptosis, but it is not clear whether PD can regulate autophagy to protect from CP-AKI. The purpose of this study was to investigate the effect of PD on autophagy in CP-treated HK-2 cells and CP-AKI mouse models, exploring the role of sirtuin 6 (SIRT6) upregulated by PD. In this study, the blocking of autophagy flux was observed in both CP-treated HK-2 cells in vitro and CP-AKI mouse models in vivo, whereas this blocking was reversed by PD, which was characterized by the increase of autophagy microtubule-associated protein light chain 3 II expression and autophagolysosome/autophagosome ratio and the decrease of p62 expression. Furthermore, PD also significantly increased the expression of SIRT6 in vivo and in vitro. The protective effect of PD manifested by the stimulating of autophagy flux, with the reducing of inflammatory response and oxidative stress, which included downregulation of tumor necrosis factor-α and interleukin-1β, decreased activity of myeloperoxidase and content of malondialdehyde, and increased activity of superoxide dismutase and level of glutathione, both in vivo and in vitro, was reversed by either inhibition of autophagy flux by chloroquine or downregulation of SIRT6 by OSS-128167. Taken together, the present findings provide the first evidence demonstrating that PD exhibited nephroprotective effects on CP-AKI by restoring SIRT6-mediated autophagy flux mechanisms.
Collapse
|