1
|
Huang Z, Li Y, Zhao Z, Ye L, Zhang T, Yu Z, Zhai E, Qian Y, Xu X, Zhao R, Cai S, Chen J. Enhanced ZBTB10 expression induced by betulinic acid inhibits gastric cancer progression by inactivating the ARRDC3/ITGB4/PI3K/AKT pathway. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01039-8. [PMID: 39873948 DOI: 10.1007/s13402-025-01039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) ranks as the fourth leading cause of cancer-related deaths worldwide, with most patients diagnosed at advanced stages due to the absence of reliable early detection biomarkers. METHODS RNA-sequencing was conducted to identify the differentially expressed genes between GC tissues and adjacent normal tissues. CCK8, EdU, colony formation, transwell, flow cytometry and xenograft assays were adopted to explore the biological function of ZBTB10 and betulinic acid (BA) in GC progression. RNA-sequencing and phospho-proteomic profiling were performed to analyze the signaling pathways associated with ZBTB10-inhibiting GC progression. Chromatin immunoprecipitation, Co-immunoprecipitation and luciferase reporter assay were employed to elucidate the potential molecular regulatory mechanisms of ZBTB10 in GC. RESULTS ZBTB10 was one of the most significantly downregulated genes in GC tissues, and higher expression levels of ZBTB10 was correlated with better prognosis in patients with GC. Functional studies revealed that ZBTB10 overexpression and BA inhibited GC progression both in vitro and in vivo. Mechanistically, ZBTB10 enhanced ARRDC3 expression by binding to a specific response element in the ARRDC3 promoter region. Elevated ARRDC3 then directly interacted with β-4 integrin (ITGB4), leading to its ubiquitination and degradation. This cascade ultimately resulted in the downregulation of PI3K and AKT phosphorylation level. Moreover, ZBTB10 was a key target for BA in GC and BA inhibited GC progression through regulating the ZBTB10/ARRDC3/ITGB4/PI3K/AKT axis. CONCLUSIONS Our findings reveal that BA holds promise as an effective therapeutic strategy for GC, and the ZBTB10/ARRDC3/ITGB4/PI3K/AKT axis may serve as a novel diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Zhixin Huang
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Zeyu Zhao
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Linying Ye
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Tianhao Zhang
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zihan Yu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ertao Zhai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yan Qian
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiang Xu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Risheng Zhao
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Shirong Cai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Jianhui Chen
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi, 530000, China.
| |
Collapse
|
2
|
Fakhredini F, Alidadi H, Mahdavinia M, Khorsandi L. Morin promotes autophagy in human PC3 prostate cancer cells by modulating AMPK/mTOR/ULK1 signaling pathway. Tissue Cell 2024; 91:102557. [PMID: 39265522 DOI: 10.1016/j.tice.2024.102557] [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: 06/23/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
AMP-activated protein kinase (AMPK) suppresses tumorigenesis by modulating autophagy and apoptosis. This study evaluated the impact of Morin on PC3 prostate cancerous cells by examining the AMPK/ mechanistic target of rapamycin (mTOR)/ ULK1 (UNC-51-like kinase 1) pathway and autophagy process. The PC3 cells were treated with Morin (50 µg/ml) and AICAR (an AMPK activator). Cell viability, apoptosis, autophagy, and level of phosphorylated and non-phosphorylated ULK1, AMPK, and mTOR, as well as LC3B/LC3A, have been investigated. Through DAPI staining, measurement of Bax/Bcl-2 ratio, Caspase activity, and Annexin V/PI method, it has been revealed that Morin induces apoptosis and reduces the growth of PC3 cells. Morin enhanced the protein level of phosphorylated AMPK (p-AMPK) and ULK1 (p-ULK1) and decreased the expression of phosphorylated mTOR (p-mTOR) in the PC3 cells. Morin could also increase the LC3B/LC3A ratio, Acridine Orange-positive cells, expression of Beclin-1 and ATG5 genes, and decrease the p62 protein level indicating autophagy-inducing. AICAR (an AMPK activator) enhanced the impact of Morin on apoptosis, cell growth, and expression of LC3B, p-AMPK, p-ULK1, and p-mTOR proteins in the PC3 cells. These findings suggest that Morin induces apoptotic and autophagic cell death by activating AMPK and ULK1 and suppressing mTOR pathways.
Collapse
Affiliation(s)
- Fereshtesadat Fakhredini
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadis Alidadi
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
3
|
Luo S, Huang X, Li S, Chen Y, Zhang X, Zeng X. Homogeneous Polyporus polysaccharide exerts anti-bladder cancer effects via autophagy induction. PHARMACEUTICAL BIOLOGY 2024; 62:214-221. [PMID: 38353262 PMCID: PMC10868468 DOI: 10.1080/13880209.2024.2316195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024]
Abstract
CONTEXT Polyporus polysaccharide (PPS), the leading bioactive ingredient extracted from Polyporus umbellatus (Pers.) Fr. (Polyporaceae), has been demonstrated to exert anti-bladder cancer and immunomodulatory functions in macrophages. OBJECTIVE To explore the effects of homogeneous Polyporus polysaccharide (HPP) on the proliferation and autophagy of bladder cancer cells co-cultured with macrophages. MATERIALS AND METHODS MB49 bladder cancer cells and RAW264.7 macrophages were co-cultured with or without HPP intervention (50, 100, or 200 μg/mL) for 24 h. The cell counting kit-8 (CCK-8) assay and 5-ethynyl-2″-deoxyuridine (EdU) staining evaluated MB49 cell proliferation. Monodansylcadaverine (MDC) staining and transmission electron microscopy (TEM) observed autophagosomes. Western blotting detected the expression levels of autophagy-related proteins and PI3K/Akt/mTOR pathway proteins. RESULTS HPP inhibited the proliferation of MB49 cells co-cultured with RAW264.7 cells but not MB49 cells alone. HPP altered the expression of autophagy-related proteins and promoted the formation of autophagosomes in MB49 cells in the co-culture system. Autophagy inhibitors 3-methyladenine (3-MA) and chloroquine (CQ) not only antagonized HPP-induced autophagy but also attenuated the inhibitory effects of HPP on MB49 cell proliferation in the co-culture system. HPP or RAW264.7 alone was not sufficient to induce autophagy in MB49 cells. In addition, HPP suppressed the protein expression of the PI3K/Akt/mTOR pathway in MB49 cells in the co-culture system. DISCUSSION AND CONCLUSIONS HPP induced bladder cancer cell autophagy by regulating macrophages in the co-culture system, resulting in the inhibition of cancer cell proliferation. The PI3K/Akt/mTOR pathway was involved in HPP-induced autophagy in the co-culture system.
Collapse
Affiliation(s)
- Siwan Luo
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaopeng Huang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shiqi Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuwen Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xian Zhang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xing Zeng
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
4
|
Liu B, Chen M, Liang Y, Mei Z, Sun W, Gao W, Zhang T, Wang R, Guo Y. ZC3H13 promotes autophagy in bladder cancer through m6A methylation modification of PJA2 and ubiquitination of KSR1. Hum Cell 2024; 38:23. [PMID: 39614918 DOI: 10.1007/s13577-024-01155-x] [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: 07/31/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
The N6-methyladenine (m6A) modification is the most common modification of messenger RNAs in eukaryotes and has crucial roles in multiple cancers, including bladder cancer (BLCA). This paper aimed to probe the molecular mechanism of zinc-finger CCCH-type containing 13 (ZC3H13)-mediated N6-methyladenine (m6A) modification in BLCA progression via autophagy. Differential expression of ZC3H13 in BLCA was analyzed by the bioinformatics database. ZC3H13 expression in BLCA tissues and cell lines was determined, and malignant behaviors of BLCA cells were examined in vitro and in vivo. ZC3H13 was decreased in BLCA tissues and cell lines relative to adjacent tissues and normal uroepithelial cells. ZC3H13 overexpression restricted BLCA cell growth in vitro and curbed BLCA development in vivo. ZC3H13 promoted the mRNA stability of paraja ring finger 2 (PJA2) through m6A modification, leading to the ubiquitination degradation of the kinase suppressor of Ras 1 (KSR1). Knockdown of PJA2 and overexpression of KSR1 reversed the inhibitory effect of ZC3H13 on BLCA progression. ZC3H13 degraded KSR1 through m6A modification of PJA2, promoted cell autophagy, and repressed BLCA progression. Overall, ZC3H13 promotes the mRNA stability of PJA2 through m6A modification to degrade KSR1, thereby promoting autophagy in BLCA.
Collapse
Affiliation(s)
- Beibei Liu
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Mengjie Chen
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Yujie Liang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Zhijie Mei
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Wei Sun
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Wuyue Gao
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Tiantian Zhang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Rui Wang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China.
| | - Yuanyuan Guo
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China.
| |
Collapse
|
5
|
Yuan Z, He J, Li Z, Fan B, Zhang L, Man X. Targeting autophagy in urological system cancers: From underlying mechanisms to therapeutic implications. Biochim Biophys Acta Rev Cancer 2024; 1879:189196. [PMID: 39426690 DOI: 10.1016/j.bbcan.2024.189196] [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: 07/29/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
The urological system, including kidneys, ureters, bladder, urethra and prostate is known to be vital for blood filtration, waste elimination and electrolyte balance. Notably, urological system cancers represent a significant portion of global cancer diagnoses and mortalities. The current therapeutic strategies for early-stage cancer primarily involve resection surgery, which significantly affects the quality of life of patients, whereas advanced-stage cancer often relies on less effective chemo- or radiotherapy. Recently, accumulating evidence has revealed that autophagy, a crucial process in which excess organelles or inclusions within cells are removed to maintain cell homeostasis, has numerous links to urological system cancers. In this review, we focus on summarizing the underlying two-sided mechanisms of autophagy in urological system cancers. We also review the current clinical drugs targeting autophagy, which demonstrate significant potential in improving treatment outcomes for urological system cancers. In addition, we provide an overview of the research status of novel small molecule compounds targeting autophagy that are in the preclinical stages of investigation. Furthermore, drug combinations based on autophagy modulation strategies in urological system cancers are systematically summarized and discussed. These findings provide comprehensive new insight for the future discovery of more autophagy-related drug candidates.
Collapse
Affiliation(s)
- Ziyue Yuan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiani He
- Department of Urology, Department of Surgical Oncology and Breast Surgery, Institute of Urology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Fan
- Department of Urology, Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Department of Urology, Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Xiaojun Man
- Department of Urology, Department of Surgical Oncology and Breast Surgery, Institute of Urology, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
6
|
Zhang Z, Gao Z, Fang H, Zhao Y, Xing R. Therapeutic importance and diagnostic function of circRNAs in urological cancers: from metastasis to drug resistance. Cancer Metastasis Rev 2024; 43:867-888. [PMID: 38252399 DOI: 10.1007/s10555-023-10152-9] [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: 06/10/2023] [Accepted: 10/31/2023] [Indexed: 01/23/2024]
Abstract
Circular RNAs (circRNAs) are a member of non-coding RNAs with no ability in encoding proteins and their aberrant dysregulation is observed in cancers. Their closed-loop structure has increased their stability, and they are reliable biomarkers for cancer diagnosis. Urological cancers have been responsible for high mortality and morbidity worldwide, and developing new strategies in their treatment, especially based on gene therapy, is of importance since these malignant diseases do not respond to conventional therapies. In the current review, three important aims are followed. At the first step, the role of circRNAs in increasing or decreasing the progression of urological cancers is discussed, and the double-edged sword function of them is also highlighted. At the second step, the interaction of circRNAs with molecular targets responsible for urological cancer progression is discussed, and their impact on molecular processes such as apoptosis, autophagy, EMT, and MMPs is highlighted. Finally, the use of circRNAs as biomarkers in the diagnosis and prognosis of urological cancer patients is discussed to translate current findings in the clinic for better treatment of patients. Furthermore, since circRNAs can be transferred to tumor via exosomes and the interactions in tumor microenvironment provided by exosomes such as between macrophages and cancer cells is of importance in cancer progression, a separate section has been devoted to the role of exosomal circRNAs in urological tumors.
Collapse
Affiliation(s)
- Zhibin Zhang
- College of Traditional Chinese Medicine, Chengde Medical College, Chengde, 067000, Hebei, China.
| | - Zhixu Gao
- Chengde Medical College, Chengde, 067000, Hebei, China
| | - Huimin Fang
- Chengde Medical College, Chengde, 067000, Hebei, China
| | - Yutang Zhao
- Chengde Medical College, Chengde, 067000, Hebei, China
| | - Rong Xing
- Chengde Medical College, Chengde, 067000, Hebei, China
| |
Collapse
|
7
|
Banerjee S, Banerjee S, Bishayee A, Da Silva MN, Sukocheva OA, Tse E, Casarcia N, Bishayee A. Cellular and molecular mechanisms underlying the potential of betulinic acid in cancer prevention and treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155858. [PMID: 39053249 DOI: 10.1016/j.phymed.2024.155858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Betulinic acid (BA), which is a pentacyclic triterpenoid found in the bark of plane, birch, and eucalyptus trees, has emerged as a compound of significant interest in scientific research due to its potential therapeutic applications. BA has a range of well-documented pharmacological and biological effects, including antibacterial, immunomodulatory, diuretic, antiviral, antiparasitic, antidiabetic, and anticancer activities. Although numerous research studies have explored the potential anticancer effects of BA, there is a noticeable gap in the literature, highlighting the need for a more up-to-date and comprehensive evaluation of BA's anticancer potential. PURPOSE The aim of this work is to critically assess the reported cellular and molecular mechanisms underlying the cancer preventive and therapeutic effects of BA. METHODS Relevant research on the inhibitory effects of BA against cancerous cells was searched using Science Direct, Scopus, Web of Science, and PubMed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS The anticancer properties of BA are mediated by the activation of cell death and cell cycle arrest, production of reactive oxygen species, increased mitochondrial permeability, modulation of nuclear factor-κB and Bcl-2 family signaling. Emerging evidence also underscores the combined anticancer effects of BA with other natural bioactive compounds or approved drugs. Notably, several novel BA nanoformulations have been found to exhibit encouraging antineoplastic activities. CONCLUSION BA, whether used alone or in combination, or as a form of nanoformulation, shows significant potential for cancer prevention and treatment. Nevertheless, further detailed studies are necessary to confirm the therapeutic effectiveness of this natural compound.
Collapse
Affiliation(s)
- Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol 713 301, West Bengal, India
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol 713 301, West Bengal, India
| | | | - Milton Nascimento Da Silva
- Laboratory of Liquid Chromatography, Institute of Exact and Natural Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; Chemistry Post-Graduation Program, Institute of Exact and Natural Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; Pharmaceutical Science Post-Graduation Program, Institute of Health Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Olga A Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Nicolette Casarcia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
8
|
Yuan JJ, Meng YF, Zhang MS, Guo DL, Yang JW, Dong MJ, Sun CX, Xiao SJ. Isoprenoid flavonoids isolated from Sophora davidii and their activities induces apoptosis and autophagy in HT29 cells. Fitoterapia 2024; 175:105945. [PMID: 38575091 DOI: 10.1016/j.fitote.2024.105945] [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: 01/21/2024] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Four previously undescribed isoprenoid flavonoids (2-5) were isolated from Sophora davidii, along with five known analogues. The structures of the compounds were established through comprehensive analysis of spectroscopic data, including HRESIMS, 1D and 2D NMR, and absolute configurations determined by theoretical calculations, including ECD and NMR calculation. The cytotoxic effects of the isolated compounds on human HT29 colon cancer cells were evaluated using the MTT assay, compound 1 exhibited cytotoxicity against human HT29 colon cancer cells with an IC50 value of 8.39 ± 0.09 μM. Studies conducted with compound 1 in HT29 cells demonstrated that it may induce apoptosis and autophagy in HT29 by promoting the phosphorylation of P38 MAPK and inhibiting the phosphorylation of Erk MAPK.
Collapse
Affiliation(s)
- Jing-Jing Yuan
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Yu-Fan Meng
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Mao-Sheng Zhang
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Da-Le Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jian-Wen Yang
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Min-Jian Dong
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Cheng-Xin Sun
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China.
| | - Shi-Ji Xiao
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
9
|
Wang H, Du X, Liu W, Zhang C, Li Y, Hou J, Yu Y, Li G, Wang Q. Combination of betulinic acid and EGFR-TKIs exerts synergistic anti-tumor effects against wild-type EGFR NSCLC by inducing autophagy-related cell death via EGFR signaling pathway. Respir Res 2024; 25:215. [PMID: 38764025 PMCID: PMC11103851 DOI: 10.1186/s12931-024-02844-9] [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: 02/18/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of lung cancer patients with mutated EGFR. However, the efficacy of EGFR-TKIs in wild-type EGFR tumors has been shown to be marginal. Methods that can sensitize EGFR-TKIs to EGFR wild-type NSCLC remain rare. Hence, we determined whether combination treatment can maximize the therapeutic efficacy of EGFR-TKIs. METHODS We established a focused drug screening system to investigate candidates for overcoming the intrinsic resistance of wild-type EGFR NSCLC to EGFR-TKIs. Molecular docking assays and western blotting were used to identify the binding mode and blocking effect of the candidate compounds. Proliferation assays, analyses of drug interactions, colony formation assays, flow cytometry and nude mice xenograft models were used to determine the effects and investigate the molecular mechanism of the combination treatment. RESULTS Betulinic acid (BA) is effective at targeting EGFR and synergizes with EGFR-TKIs (gefitinib and osimertinib) preferentially against wild-type EGFR. BA showed inhibitory activity due to its interaction with the ATP-binding pocket of EGFR and dramatically enhanced the suppressive effects of EGFR-TKIs by blocking EGFR and modulating the EGFR-ATK-mTOR axis. Mechanistic studies revealed that the combination strategy activated EGFR-induced autophagic cell death and that the EGFR-AKT-mTOR signaling pathway was essential for completing autophagy and cell cycle arrest. Activation of the mTOR pathway or blockade of autophagy by specific chemical agents markedly attenuated the effect of cell cycle arrest. In vivo administration of the combination treatment caused marked tumor regression in the A549 xenografts. CONCLUSIONS BA is a potential wild-type EGFR inhibitor that plays a critical role in sensitizing EGFR-TKI activity. BA combined with an EGFR-TKI effectively suppressed the proliferation and survival of intrinsically resistant lung cancer cells via the inhibition of EGFR as well as the induction of autophagy-related cell death, indicating that BA combined with an EGFR-TKI may be a potential therapeutic strategy for overcoming the primary resistance of wild-type EGFR-positive lung cancers.
Collapse
Affiliation(s)
- Han Wang
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
- Guangzhou women and children's medical center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaohui Du
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Wenwen Liu
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Congcong Zhang
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Ying Li
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Jingwen Hou
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Yi Yu
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Guiru Li
- The Second Hospital of Dalian Medical University, Dalian, 116023, China.
| | - Qi Wang
- The Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
10
|
Shen W, Zeng X, Zeng X, Hu B, Ren C, Lin Z, Zhang L, Rui G, Yasen M, Chen X. Trifluoperazine activates AMPK / mTOR / ULK1 signaling pathway to induce mitophagy in osteosarcoma cells. Chem Biol Interact 2024; 392:110904. [PMID: 38360085 DOI: 10.1016/j.cbi.2024.110904] [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: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
Osteosarcoma is a prevalent kind of primary bone malignancy. Trifluoperazine, as an antipsychotic drug, has anti-tumor activity against a variety of cancers. Nevertheless, the impact of trifluoperazine on osteosarcoma is unclear. Our investigation aimed to explore the mechanism of trifluoperazine's effect on osteosarcoma. We found that trifluoperazine inhibited 143B and U2-OS osteosarcoma cell proliferation in a method based on the dose. Furthermore, it was shown that trifluoperazine induced the accumulation of reactive oxygen species (ROS) to cause mitochondrial damage and induced mitophagy in osteosarcoma cells. Finally, combined with RNA-seq results, we first demonstrated the AMPK/mTOR/ULK1 signaling pathway as a potential mechanism of trifluoperazine-mediated mitophagy in osteosarcoma cells and can be suppressed by AMPK inhibitor Compound C.
Collapse
Affiliation(s)
- Wenhui Shen
- Department of Orthopedic Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, China
| | - Xiangchen Zeng
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Xianhui Zeng
- Department of Infectious Diseases, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, 570206, China
| | - Baoshan Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Chong Ren
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Zhiming Lin
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Long Zhang
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Gang Rui
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China
| | - Miersalijiang Yasen
- Department of Orthopedic Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361000, China.
| | - Xiaohui Chen
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, 361001, China.
| |
Collapse
|
11
|
Fernandes S, Vieira M, Prudêncio C, Ferraz R. Betulinic Acid for Glioblastoma Treatment: Reality, Challenges and Perspectives. Int J Mol Sci 2024; 25:2108. [PMID: 38396785 PMCID: PMC10889789 DOI: 10.3390/ijms25042108] [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: 01/03/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Betulinic acid is a naturally occurring compound that can be obtained through methanolic or ethanolic extraction from plant sources, as well as through chemical synthesis or microbial biotransformation. Betulinic acid has been investigated for its potential therapeutic properties, and exhibits anti-inflammatory, antiviral, antimalarial, and antioxidant activities. Notably, its ability to cross the blood-brain barrier addresses a significant challenge in treating neurological pathologies. This review aims to compile information about the impact of betulinic acid as an antitumor agent, particularly in the context of glioblastoma. Importantly, betulinic acid demonstrates selective antitumor activity against glioblastoma cells by inhibiting proliferation and inducing apoptosis, consistent with observations in other cancer types. Compelling evidence published highlights the acid's therapeutic action in suppressing the Akt/NFκB-p65 signaling cascade and enhancing the cytotoxic effects of the chemotherapeutic agent temozolomide. Interesting findings with betulinic acid also suggest a focus on researching the reduction of glioblastoma's invasiveness and aggressiveness profile. This involves modulation of extracellular matrix components, remodeling of the cytoskeleton, and secretion of proteolytic proteins. Drawing from a comprehensive review, we conclude that betulinic acid formulations as nanoparticles and/or ionic liquids are promising drug delivery approaches with the potential for translation into clinical applications for the treatment and management of glioblastoma.
Collapse
Affiliation(s)
- Sílvia Fernandes
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic University of Porto, Rua Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal; (S.F.); (C.P.)
- Center for Research on Health and Environment (CISA), School of Health (ESS), Polytechnic University of Porto, Rua Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal
| | - Mariana Vieira
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic University of Porto, Rua Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal; (S.F.); (C.P.)
| | - Cristina Prudêncio
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic University of Porto, Rua Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal; (S.F.); (C.P.)
- Ciências Químicas e das Biomoléculas, School of Health (ESS), Polytechnic University of Porto, Rua Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal
| | - Ricardo Ferraz
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic University of Porto, Rua Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal; (S.F.); (C.P.)
- Ciências Químicas e das Biomoléculas, School of Health (ESS), Polytechnic University of Porto, Rua Dr. António Bernardino de Almeida, 400, 4200-072 Porto, Portugal
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| |
Collapse
|
12
|
Wang C, Yu J, Ding C, Chen C. CangFu Daotan decoction improves polycystic ovarian syndrome by downregulating FOXK1. Gynecol Endocrinol 2023; 39:2244600. [PMID: 37544927 DOI: 10.1080/09513590.2023.2244600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/09/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
Objective: Polycystic ovarian syndrome (PCOS) is a prevalent gynecologic disorder, often associated with abnormal follicular development. Cangfu Daotan decoction (CFD) is a traditional Chinese medicine formula that is effective in alleviating PCOS clinically, but the specific mechanism remains unclear. Forkhead box K1 (FOXK1) is associated with cellular function. This study aimed to explore the effects of CFD and FOXK1 on PCOS.Methods: High-fat diet and letrozole were combined to establish PCOS rat models. Next, primary GCs were extracted from those PCOS rats. Then, GC cells were transfected with si-FOXK1 or oe-FOXK1. CFD-contain serum was prepared, and experiments were conducted to investigate the regulation of FOXK1 by CFD.Results: FOXK1 was highly expressed in GCs of PCOS rats. Further investigation revealed that FOXK1 overexpression resulted in inhibition of proliferation and DNA synthesis, along with promotion of apoptosis and autophagy in GCs. Additionally, it was found that FOXK1 promoted the expressions of the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway-related proteins. Interestingly, treatment with CFD reversed all the effects of FOXK1 overexpression in GCs. Conclusion: This study demonstrated that CFD exerted a protective role in PCOS by inhibiting FOXK1, which provided a research basis for the application of CFD in PCOS, and suggested that FOXK1 is a novel therapeutic target in PCOS treatment.
Collapse
Affiliation(s)
- Chenye Wang
- Department of Reproductive Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou City, China
| | - Jia Yu
- Department of Reproductive Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou City, China
| | - Caifei Ding
- Department of Reproductive Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou City, China
| | - Chunyue Chen
- Department of Reproductive Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou City, China
| |
Collapse
|
13
|
Willig JB, de Couto NMG, Vianna DRB, Mariot CDS, Gnoatto SCB, Buffon A, Pilger DA. Betulinic Acid-Brosimine B Hybrid Compound Has a Synergistic Effect with Imatinib in Chronic Myeloid Leukemia Cell Line, Modulating Apoptosis and Autophagy. Pharmaceuticals (Basel) 2023; 16:ph16040586. [PMID: 37111343 PMCID: PMC10142704 DOI: 10.3390/ph16040586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease characterized by the formation of the BCR-ABL (breakpoint cluster region-Abelson) oncoprotein. As many patients display therapeutic resistance, the development of new drugs based on semisynthetic products represents a new potential therapeutic approach for treating the disease. In this study, we investigated the cytotoxic activity, possible mechanism of action of a hybrid compound of betulinic acid (BA) and brosimine B in CML cell lines that are sensitive (K-562) and resistant (K-562R) to imatinib, in addition to evaluating lower doses of imatinib in combination with the hybrid compound. The effects of the compound, and its combination with imatinib, on apoptosis, cell cycle, autophagy and oxidative stress were determined. The compound was cytotoxic in K-562 (23.57 ± 2.87 μM) and K-562R (25.80 ± 3.21 μM) cells, and a synergistic effect was observed when it was associated with imatinib. Apoptosis was mediated by the caspase 3 and 9 intrinsic pathway, and cell cycle evaluation showed arrest at G0/G1. In addition, the hybrid compound increased the production of reactive oxygen species and induced autophagy by increasing LC3II and Beclin-1 mRNA levels. Results suggest that this hybrid compound causes the death of both imatinib-sensitive and -resistant cell lines and may hold potential as a new anticancer treatment against CML.
Collapse
Affiliation(s)
- Julia Biz Willig
- Post-Graduation of Pharmaceutical Science Program, Faculty of Farmacy, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
- Laboratory Biochemical and Cytological Analysis, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
| | - Nádia Miléo Garcês de Couto
- Post-Graduation of Pharmaceutical Science Program, Faculty of Farmacy, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
- Laboratory of Phytochemistry and Organic Synthesis, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
| | - Débora Renz Barreto Vianna
- Post-Graduation of Pharmaceutical Science Program, Faculty of Farmacy, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
- Laboratory Biochemical and Cytological Analysis, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
| | - Camila da Silveira Mariot
- Laboratory Biochemical and Cytological Analysis, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
| | - Simone Cristina Baggio Gnoatto
- Post-Graduation of Pharmaceutical Science Program, Faculty of Farmacy, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
- Laboratory of Phytochemistry and Organic Synthesis, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
| | - Andréia Buffon
- Laboratory Biochemical and Cytological Analysis, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
| | - Diogo André Pilger
- Post-Graduation of Pharmaceutical Science Program, Faculty of Farmacy, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
- Laboratory Biochemical and Cytological Analysis, Federal University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil
| |
Collapse
|
14
|
The role of BMI1 in endometrial cancer and other cancers. Gene 2023; 856:147129. [PMID: 36563713 DOI: 10.1016/j.gene.2022.147129] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Endometrial cancer (EC) is the third leading gynecological malignancy, and its treatment remains challenging. B cell-specific Moloney murine leukemia virus integration site-1 (BMI1) is one of the core members of the polycomb group (PcG) family, which plays a promoting role in the occurrence and development of various tumors. Notably, BMI1 has been found to be frequently upregulated in endometrial cancer (EC) and promote the occurrence of EC through promoting epithelial-mesenchymal transition (EMT) and AKT/PI3K pathways. This review summarizes the structure and upstream regulatory mechanisms of BMI1 and its role in EC. In addition, we focused on the role of BMI1 in chemoradiotherapy resistance and summarized the current drugs that target BMI1.
Collapse
|
15
|
Yang L, Nao J. Focus on Alzheimer's Disease: The Role of Fibroblast Growth Factor 21 and Autophagy. Neuroscience 2023; 511:13-28. [PMID: 36372296 DOI: 10.1016/j.neuroscience.2022.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/24/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
Alzheimer's disease (AD) is a disorder of the central nervous system that is typically marked by progressive cognitive impairment and memory loss. Amyloid β plaque deposition and neurofibrillary tangles with hyperphosphorylated tau are the two hallmark pathologies of AD. In mammalian cells, autophagy clears aberrant protein aggregates, thus maintaining proteostasis as well as neuronal health. Autophagy affects production and metabolism of amyloid β and accumulation of phosphorylated tau proteins, whose malfunction can lead to the progression of AD. On the other hand, defective autophagy has been found to induce the production of the neuroprotective factor fibroblast growth factor 21 (FGF21), although the underlying mechanism is unclear. In this review, we highlight the significance of aberrant autophagy in the pathogenesis of AD, discuss the possible mechanisms by which defective autophagy induces FGF21 production, and analyze the potential of FGF21 in the treatment of AD. The findings provide some insights into the potential role of FGF21 and autophagy in the pathogenesis of AD.
Collapse
Affiliation(s)
- Lan Yang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
16
|
Yang Y, Chen Y, Wu JH, Ren Y, Liu B, Zhang Y, Yu H. Targeting regulated cell death with plant natural compounds for cancer therapy: A revisited review of apoptosis, autophagy-dependent cell death, and necroptosis. Phytother Res 2023; 37:1488-1525. [PMID: 36717200 DOI: 10.1002/ptr.7738] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 02/01/2023]
Abstract
Regulated cell death (RCD) refers to programmed cell death regulated by various protein molecules, such as apoptosis, autophagy-dependent cell death, and necroptosis. Accumulating evidence has recently revealed that RCD subroutines have several links to many types of human cancer; therefore, targeting RCD with pharmacological small-molecule compounds would be a promising therapeutic strategy. Moreover, plant natural compounds, small-molecule compounds synthesized from plant sources, and their derivatives have been widely reported to regulate different RCD subroutines to improve potential cancer therapy. Thus, in this review, we focus on updating the intricate mechanisms of apoptosis, autophagy-dependent cell death, and necroptosis in cancer. Moreover, we further discuss several representative plant natural compounds and their derivatives that regulate the above-mentioned three subroutines of RCD, and their potential as candidate small-molecule drugs for the future cancer treatment.
Collapse
Affiliation(s)
- Yuanyuan Yang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanmei Chen
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jun Hao Wu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yueting Ren
- Department of Pharmacology and Toxicology, Temerity Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
17
|
Dong L, He J, Luo L, Wang K. Targeting the Interplay of Autophagy and ROS for Cancer Therapy: An Updated Overview on Phytochemicals. Pharmaceuticals (Basel) 2023; 16:ph16010092. [PMID: 36678588 PMCID: PMC9865312 DOI: 10.3390/ph16010092] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Autophagy is an evolutionarily conserved self-degradation system that recycles cellular components and damaged organelles, which is critical for the maintenance of cellular homeostasis. Intracellular reactive oxygen species (ROS) are short-lived molecules containing unpaired electrons that are formed by the partial reduction of molecular oxygen. It is widely known that autophagy and ROS can regulate each other to influence the progression of cancer. Recently, due to the wide potent anti-cancer effects with minimal side effects, phytochemicals, especially those that can modulate ROS and autophagy, have attracted great interest of researchers. In this review, we afford an overview of the complex regulatory relationship between autophagy and ROS in cancer, with an emphasis on phytochemicals that regulate ROS and autophagy for cancer therapy. We also discuss the effects of ROS/autophagy inhibitors on the anti-cancer effects of phytochemicals, and the challenges associated with harnessing the regulation potential on ROS and autophagy of phytochemicals for cancer therapy.
Collapse
Affiliation(s)
- Lixia Dong
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Jingqiu He
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Li Luo
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
- Correspondence: (L.L.); (K.W.)
| | - Kui Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
- Correspondence: (L.L.); (K.W.)
| |
Collapse
|
18
|
Farooqi AA, Turgambayeva A, Tashenova G, Tulebayeva A, Bazarbayeva A, Kapanova G, Abzaliyeva S. Multifunctional Roles of Betulinic Acid in Cancer Chemoprevention: Spotlight on JAK/STAT, VEGF, EGF/EGFR, TRAIL/TRAIL-R, AKT/mTOR and Non-Coding RNAs in the Inhibition of Carcinogenesis and Metastasis. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010067. [PMID: 36615262 PMCID: PMC9822120 DOI: 10.3390/molecules28010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
The pursual of novel anticancer molecules from natural sources has gained worthwhile appreciation, and a significant fraction of conceptual knowledge has revolutionized our understanding about heterogeneous nature of cancer. Betulinic acid has fascinated interdisciplinary researchers due to its tremendous pharmacological properties. Ground-breaking discoveries have unraveled previously unprecedented empirical proof-of-concept about momentous chemopreventive role of betulinic acid against carcinogenesis and metastasis. Deregulation of cell signaling pathways has been reported to play a linchpin role in cancer progression and colonization of metastatically competent cancer cells to the distant organs for the development of secondary tumors. Importantly, betulinic acid has demonstrated unique properties to mechanistically modulate oncogenic transduction cascades. In this mini-review, we have attempted to provide a sophisticated compendium of regulatory role of betulinic acid in cancer chemoprevention. We have partitioned this multi-component review into different sections in which we summarized landmark research-works which highlighted betulinic acid mediated regulation of JAK/STAT, VEGF, EGF/EGFR, TRAIL/TRAIL-R, AKT/mTOR and ubiquitination pathways in the inhibition of cancer. In parallel, betulinic acid mediated regulation of signaling cascades and non-coding RNAs will be critically analyzed in cell culture and animal model studies. Better comprehension of the pharmaceutical features of betulinic acid and mapping of the existing knowledge gaps will be valuable in the translatability of preclinical studies into rationally designed clinical trials.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
- Correspondence:
| | - Assiya Turgambayeva
- Department of Public Health and Management, NJSC “Astana Medical University”, Astana 010000, Kazakhstan
| | - Gulnara Tashenova
- Asfendiyarov Kazakh National Medical University, Almaty 050040, Kazakhstan
- Scientific Center of Pediatrics and Pediatric Surgery, Almaty 050060, Kazakhstan
| | - Aigul Tulebayeva
- Asfendiyarov Kazakh National Medical University, Almaty 050040, Kazakhstan
- Scientific Center of Pediatrics and Pediatric Surgery, Almaty 050060, Kazakhstan
| | - Aigul Bazarbayeva
- Scientific Center of Pediatrics and Pediatric Surgery, Almaty 050060, Kazakhstan
| | - Gulnara Kapanova
- Scientific Center of Anti-Infectious Drugs, 75 al-Faraby Ave, Almaty 050040, Kazakhstan
- Al-Farabi Kazakh National University, 71 al-Farabi Ave, Almaty 050040, Kazakhstan
| | - Symbat Abzaliyeva
- Al-Farabi Kazakh National University, 71 al-Farabi Ave, Almaty 050040, Kazakhstan
| |
Collapse
|
19
|
Crosstalk of miRNAs with signaling networks in bladder cancer progression: Therapeutic, diagnostic and prognostic functions. Pharmacol Res 2022; 185:106475. [DOI: 10.1016/j.phrs.2022.106475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 09/27/2022] [Indexed: 12/24/2022]
|
20
|
Ataş MN, Ertuğrul B, İplik ES, Çakmakoğlu B, Ergen A. The inhibitory effect of betulinic acid on epithelial-mesenchymal transition pathway in renal cell carcinoma. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:170. [PMID: 35972672 DOI: 10.1007/s12032-022-01775-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022]
Abstract
Renal cancer is the most lethal urological cancer and characterized by high metastasis rate at initial diagnosis and drug resistance to current chemotherapeutics. Betulinic acid is a pentacyclic triterpene with broad biological activity that occurs naturally in variety of plants. Even though the anti-cancer efficacy of betulinic acid have been reported by many studies, the information about the pathways and the molecules which are affected by betulinic acid in renal cancer are limited. Epithelial-mesenchymal transition (EMT) is considered as the initial step of metastasis and contributes to drug resistance of cancer cells. Depending on the role of EMT in cancer progression and drug resistance, targeting EMT may represent an effective strategy in this context. Therefore, we aimed to investigate the anti-metastatic effects of betulinic acid on renal cell carcinoma cells by evaluating two EMT markers, SNAIL-1, and SDC-2. Following the treatment of betulinic acid at determined doses by WST-1 cytotoxicity assay in our previous study, SDC-2 expression level was decreased in both cell lines. Additionally, in correlation with this result, we also found a reduction in SDC-2 and SNAIL-1 protein levels which are measured by ELISA. Furthermore, the migration and invasion capacities were suppressed by betulinic acid treatment in metastatic renal adenocarcinoma ACHN cells. Taken together, our findings indicate that betulinic acid may constitute a potential treatment approach for renal cancer with further investigations.
Collapse
Affiliation(s)
- Merve Nur Ataş
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.,Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Barış Ertuğrul
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Elif Sinem İplik
- Department of Biochemistry, Faculty of Medicine, Istanbul Health and Technology University, Istanbul, Turkey
| | - Bedia Çakmakoğlu
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Arzu Ergen
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
21
|
Recent Advances Regarding the Molecular Mechanisms of Triterpenic Acids: A Review (Part II). Int J Mol Sci 2022; 23:ijms23168896. [PMID: 36012159 PMCID: PMC9408012 DOI: 10.3390/ijms23168896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 12/18/2022] Open
Abstract
Triterpenic acids are a widespread class of phytocompounds which have been found to possess valuable therapeutic properties such as anticancer, anti-inflammatory, hepatoprotective, cardioprotective, antidiabetic, neuroprotective, lipolytic, antiviral, and antiparasitic effects. They are a subclass of triterpenes bearing a characteristic lipophilic structure that imprints unfavorable in vivo properties which subsequently limit their applications. The early investigation of the mechanism of action (MOA) of a drug candidate can provide valuable information regarding the possible side effects and drug interactions that may occur after administration. The current paper aimed to summarize the most recent (last 5 years) studies regarding the MOA of betulinic acid, boswellic acid, glycyrrhetinic acid, madecassic acid, moronic acid, and pomolic acid in order to provide scientists with updated and accessible material on the topic that could contribute to the development of future studies; the paper stands as the sequel of our previously published paper regarding the MOA of triterpenic acids with therapeutic value. The recent literature published on the topic has highlighted the role of triterpenic acids in several signaling pathways including PI3/AKT/mTOR, TNF-alpha/NF-kappa B, JNK-p38, HIF-α/AMPK, and Grb2/Sos/Ras/MAPK, which trigger their various biological activities.
Collapse
|
22
|
Li H, Pang B, Nie B, Qu S, Zhang K, Xu J, Yang M, Liu J, Li S. Dioscin promotes autophagy by regulating the AMPK-mTOR pathway in ulcerative colitis. Immunopharmacol Immunotoxicol 2022; 44:238-246. [PMID: 35174751 DOI: 10.1080/08923973.2022.2037632] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dioscin is reported to alleviate the dextran sodium sulfate (DSS)-induced ulcerative colitis (UC) in mice. Autophagy plays an anti-inflammatory role in UC. We herein aimed to explore the biological functions of dioscin in autophagy in UC. METHODS To explore the effects of dioscin on UC progression, a DSS-induced mouse model of UC was established. Body weight, disease activity index and macroscopic damage index scores were recorded for seven days. Hematoxylin & Eosin (HE) staining was used to stain colon sections and an BX53 microscope was prepared to observe pathological changes. The activities of glutathione, superoxidative dismutase, and malondialdehyde were determined by commercially available kits. Western blotting was performed to measure the protein levels of p-AMPK/AMPK, p-mTOR/mTOR and autophagy-related genes. RESULTS The DSS-induced colitis and oxidative stress in mice were ameliorated after dioscin treatment. Dioscin promoted the phosphorylation of AMPK to inhibit mTOR activation and facilitated autophagy in DSS-induced mice model of UC. CONCLUSION Dioscin promotes autophagy by promoting the phosphorylation of AMPK to inhibit mTOR activation in ulcerative colitis.
Collapse
Affiliation(s)
- Han Li
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Bo Pang
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Bin Nie
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Shifang Qu
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Kuanxin Zhang
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Jinxiu Xu
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Ming Yang
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Jie Liu
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Shasha Li
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| |
Collapse
|
23
|
Lee CW, Huang CCY, Chi MC, Lee KH, Peng KT, Fang ML, Chiang YC, Liu JF. Naringenin Induces ROS-Mediated ER Stress, Autophagy, and Apoptosis in Human Osteosarcoma Cell Lines. Molecules 2022; 27:373. [PMID: 35056691 PMCID: PMC8781290 DOI: 10.3390/molecules27020373] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/25/2021] [Accepted: 12/28/2021] [Indexed: 11/17/2022] Open
Abstract
Osteosarcoma, a primary bone tumor, responds poorly to chemotherapy and radiation therapy in children and young adults; hence, as the basis for an alternative treatment, this study investigated the cytotoxic and antiproliferative effects of naringenin on osteosarcoma cell lines, HOS and U2OS, by using cell counting kit-8 and colony formation assays. DNA fragmentation and the increase in the G2/M phase in HOS and U2OS cells upon treatment with various naringenin concentrations were determined by using the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay and Annexin V/propidium iodide double staining, respectively. Flow cytometry was performed, and 2',7'-dichlorodihydrofluorescein diacetate, JC-1, and Fluo-4 AM ester probes were examined for reactive oxygen species (ROS) generation, mitochondrial membrane potential, and intracellular calcium levels, respectively. Caspase activation, cell cycle, cytosolic and mitochondrial, and autophagy-related proteins were determined using western blotting. The results indicated that naringenin significantly inhibited viability and proliferation of osteosarcoma cells in a dose-dependent manner. In addition, naringenin induced cell cycle arrest in osteosarcoma cells by inhibiting cyclin B1 and cyclin-dependent kinase 1 expression and upregulating p21 expression. Furthermore, naringenin significantly inhibited the growth of osteosarcoma cells by increasing the intracellular ROS level. Naringenin induced endoplasmic reticulum (ER) stress-mediated apoptosis through the upregulation of ER stress markers, GRP78 and GRP94. Naringenin caused acidic vesicular organelle formation and increased autophagolysosomes, microtubule-associated protein-light chain 3-II protein levels, and autophagy. The findings suggest that the induction of cell apoptosis, cell cycle arrest, and autophagy by naringenin through mitochondrial dysfunction, ROS production, and ER stress signaling pathways contribute to the antiproliferative effect of naringenin on osteosarcoma cells.
Collapse
Affiliation(s)
- Chiang-Wen Lee
- Department of Nursing, Division of Basic Medical Sciences, Chronic Diseases and Health Promotion Research Center and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Puzi City 61363, Taiwan;
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City 61363, Taiwan;
- Department of Safety Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City 243303, Taiwan
| | - Cathy Chia-Yu Huang
- Department of Life Sciences, National Central University, Taoyuan City 320317, Taiwan;
| | - Miao-Ching Chi
- Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City 613, Taiwan;
| | - Kuan-Han Lee
- Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Kuo-Ti Peng
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City 61363, Taiwan;
| | - Mei-Ling Fang
- Center for Environmental Toxin and Emerging-Contaminant Research, Cheng Shiu University, Kaohsiung 833, Taiwan;
- Super Micro Research and Technology Center, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Yao-Chang Chiang
- Department of Nursing, Division of Basic Medical Sciences, Chronic Diseases and Health Promotion Research Center and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Puzi City 61363, Taiwan;
| | - Ju-Fang Liu
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
24
|
Luo X, Bao X, Weng X, Bai X, Feng Y, Huang J, Liu S, Jia H, Yu B. The protective effect of quercetin on macrophage pyroptosis via TLR2/Myd88/NF-κB and ROS/AMPK pathway. Life Sci 2021; 291:120064. [PMID: 34688696 DOI: 10.1016/j.lfs.2021.120064] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/05/2021] [Accepted: 10/14/2021] [Indexed: 12/15/2022]
Abstract
AIMS Pyroptosis is a pro-inflammatory form of programmed cell death, which plays a vital role in the development of inflammatory diseases. As a natural flavonoid, quercetin has been shown to possess anti-inflammatory activity, but its effects on macrophage pyroptosis is still unclear. Therefore, this study aims to investigate the effects of quercetin on macrophage pyroptosis and the underlying mechanism. MATERIAL AND METHODS LPS/ATP treatment was used to induce THP-1 macrophage pyroptosis. Cell counting kit-8 (CCK-8) assay was used to evaluate cell viability. Scanning electron microscope (SEM) was used to detect cell morphology. Hoechst/propidium iodide (PI) staining and lactate dehydrogenase (LDH) assay were performed to evaluate the cell membrane integrity. The expression of key components and effectors of nod-like receptors3 (NLRP3) inflammasome were examined by real-time PCR and western blot. Immunofluorescence staining was used to detect reactive oxygen species (ROS) level and P65 nuclear translocation. KEY FINDINGS Our results showed that quercetin prevented THP-1 macrophage pyroptosis by reducing the expression of NLRP3 and cleaved-caspase1, as well as IL-1β and N-GSDMD in a concentration dependent manner. Quercetin suppressed NLRP3 inflammasome activation by inhibiting ROS overproduction. Moreover, quercetin inhibited the phosphorylation of P65 and its translocation from cytoplasm into nuclear. In addition, we found that quercetin suppressed the increase of TLR2/Myd88 and p-AMPK induced by LPS/ATP, while both TLR2 and AMPK agonist weakened the inhibitory effect of quercetin on the activity of NLRP3 inflammasome and alleviated the protective effect on macrophages pyroptosis. SIGNIFICANCE Quercetin possesses a protective effect on macrophages pyroptosis via TLR2/Myd88/NF-κB and ROS/AMPK pathway.
Collapse
Affiliation(s)
- Xing Luo
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Xiaoyi Bao
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Xiuzhu Weng
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Xiaoxuan Bai
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Yi Feng
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Jianxin Huang
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| | - Shaoyu Liu
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China; Bin xian People's Hospital, Harbin 150400, PR China
| | - Haibo Jia
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China.
| | - Bo Yu
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin 150001, PR China
| |
Collapse
|