1
|
Xu X, Wang Y, Yan D, Ren C, Cai Y, Liao S, Kong L, Han C. Enhanced Fe(II)-artemisinin-mediated chemodynamic therapy with efficient Fe(III)/Fe(II) conversion circulation for cancer treatment. Biomater Sci 2024. [PMID: 39422664 DOI: 10.1039/d4bm01095g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Existing strategies to investigate the antitumor effects of artemisinin and its derivatives (ART) are inadequate. Both free Fe(II) and heme in mitochondria have been proposed to be ART activators. However, the two impact factors have been considered separately or have not been thoroughly investigated. Here, the designed ART-based novel nanosystem with transferrin-modified hollow mesoporous silica nanoparticles as drug-delivery carriers is loaded with a functional artemisinin derivative (Cou-DHA), glucose oxidase, and perfluoropentane inside the cavity, which can enhance synergistic Fe(II)-ART-mediated chemodynamic therapy (CDT). Under the action of H2O2 generated by starvation therapy, the Fenton reaction occurs with Fe(III) in transferrin converted into free Fe(II). Remarkably, this report is the first to provide Fe(II) to ART actively and efficiently by combining starvation therapy and Fenton reaction-based CDT. Importantly, mitochondria-targeted Cou-DHA delivers ART into the mitochondria to sensitize the anticancer effects of ART with the supplied Fe(II) to realize Fe(II)-ART-mediated CDT. The ART-based novel nanosystem developed in our work thus has great potential for exploitation in advanced cancer therapies.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, PR China
| | - Yun Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, Center for Analysis and Testing, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Dan Yan
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, Center for Analysis and Testing, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Chunling Ren
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, Center for Analysis and Testing, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yuqian Cai
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, Center for Analysis and Testing, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Shanting Liao
- Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, PR China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, Center for Analysis and Testing, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Chao Han
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, Center for Analysis and Testing, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| |
Collapse
|
2
|
Li T, Zhang K, Gong X, Wang Q, He H, Li J, Tu P, Song Y. Online ligand screening for cytochrome C from herbal medicines through "four-in-one" measurement. Pharmacol Res 2024; 209:107406. [PMID: 39278298 DOI: 10.1016/j.phrs.2024.107406] [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: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Affinity-oriented online ligand screening with LC coupled to different detectors is widely popular to capture active compounds from herbal medicines (HMs). However, false-positive extensively occurs because insufficient information is recorded for the existence and stability of ligand-protein complex. Here, efforts were made to advance the hit confidences via configuring post-column infusion-LC-energy-resolved-affinity MS (PCI-LC-ER-AMS) to achieve "four-in-one" monitoring of: 1) response decrement of potential ligands; 2) response decrement of protein; 3) ions relating to ligand-protein complexes; and 4) ligand-protein binding strength. Ligand fishing for Cyt C from HMs was conducted as a proof-of-concept. For utility justification, a mimic sample containing twelve well-defined ligands and two negative controls underwent LC separation and met Cyt C prior to Qtof-MS measurements. Compared to Cyt C- or ligand-free assay, twelve ligands instead of negative controls showed response decrements that were consistent with twelve negative peaks observed at retention times corresponding to the ligands in Cyt C ion current chromatogram. Serial ions correlating to each ligand-Cyt C complex were observed. After recording breakdown graphs, optimal collision energy (OCE) corresponding to the non-covalent bond dissociation was positively correlated with binding strength. Two HMs including Scutellariae Radix (SR) and Aconiti Lateralis Radix Preparata were investigated. Consequently, 24 compounds were merely fished from SR, and particularly, flavonoid glycosides exhibited greater OCEs and also binding strengths over aglycones. Affinity assays and cellular evaluations consolidated the significant interactions between each captured compound and Cyt C. Overall, PCI-LC-ER-AMS is eligible for confidence-enhanced online ligand screening for Cyt C from HMs through "four-in-one" measurement.
Collapse
Affiliation(s)
- Ting Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ke Zhang
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingcheng Gong
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qian Wang
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hangyun He
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yuelin Song
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
3
|
Feng Z, Luan M, Zhu W, Xing Y, Ma X, Wang Y, Jia Y. Targeted ferritinophagy in gastrointestinal cancer: from molecular mechanisms to implications. Arch Toxicol 2024; 98:2007-2018. [PMID: 38602537 DOI: 10.1007/s00204-024-03745-y] [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: 02/27/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Gastrointestinal cancer is a significant global health burden, necessitating the development of novel therapeutic strategies. Emerging evidence has highlighted the potential of targeting ferritinophagy as a promising approach for the treatment of gastrointestinal cancer. Ferritinophagy is a form of selective autophagy that is mediated by the nuclear receptor coactivator 4 (NCOA4). This process plays a crucial role in regulating cellular iron homeostasis and has been implicated in various pathological conditions, including cancer. This review discusses the molecular mechanisms underlying ferritinophagy and its relevance to gastrointestinal cancer. Furthermore, we highlight the potential therapeutic implications of targeting ferritinophagy in gastrointestinal cancer. Several approaches have been proposed to modulate ferritinophagy, including small molecule inhibitors and immunotherapeutic strategies. We discuss the advantages and challenges associated with these therapeutic interventions and provide insights into their potential clinical applications.
Collapse
Affiliation(s)
- Zhaotian Feng
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China
| | - Yanfei Jia
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China.
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, 250013, People's Republic of China.
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, People's Republic of China.
| |
Collapse
|
4
|
Műzes G, Sipos F. Inflammasomes Are Influenced by Epigenetic and Autophagy Mechanisms in Colorectal Cancer Signaling. Int J Mol Sci 2024; 25:6167. [PMID: 38892354 PMCID: PMC11173330 DOI: 10.3390/ijms25116167] [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: 04/28/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Inflammasomes contribute to colorectal cancer signaling by primarily inducing inflammation in the surrounding tumor microenvironment. Its role in inflammation is receiving increasing attention, as inflammation has a protumor effect in addition to inducing tissue damage. The inflammasome's function is complex and controlled by several layers of regulation. Epigenetic processes impact the functioning or manifestation of genes that are involved in the control of inflammasomes or the subsequent signaling cascades. Researchers have intensively studied the significance of epigenetic mechanisms in regulation, as they encompass several potential therapeutic targets. The regulatory interactions between the inflammasome and autophagy are intricate, exhibiting both advantageous and harmful consequences. The regulatory aspects between the two entities also encompass several therapeutic targets. The relationship between the activation of the inflammasome, autophagy, and epigenetic alterations in CRC is complex and involves several interrelated pathways. This article provides a brief summary of the newest studies on how epigenetics and autophagy control the inflammasome, with a special focus on their role in colorectal cancer. Based on the latest findings, we also provide an overview of the latest therapeutic ideas for this complex network.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
5
|
Zamarioli LDS, Santos MRM, Erustes AG, Meccatti VM, Pereira TC, Smaili SS, Marcucci MC, Oliveira CR, Pereira GJS, Bincoletto C. Artemisia vulgaris Induces Tumor-Selective Ferroptosis and Necroptosis via Lysosomal Ca 2+ Signaling. Chin J Integr Med 2024; 30:525-533. [PMID: 38040876 DOI: 10.1007/s11655-023-3712-2] [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: 04/26/2023] [Indexed: 12/03/2023]
Abstract
OBJECTIVE To evaluate the chemical composition and effects of Artemisia vulgaris (AV) hydroalcoholic extract (HEAV) on breast cancer cells (MCF-7 and SKBR-3), chronic myeloid leukemia (K562) and NIH/3T3 fibroblasts. METHODS Phytochemical analysis of HEAV was done by high-performance liquid chromatography-mass (HPLC) spectrometry. Viability and cell death studies were performed using trypan blue and Annexin/FITC-7AAD, respectively. Ferrostatin-1 (Fer-1) and necrostatin-1 (Nec-1) were used to assess the mode of HEAV-induced cell death and acetoxymethylester (BAPTA-AM) was used to verify the involvement of cytosolic calcium in this event. Cytosolic calcium measurements were made using Fura-2-AM. RESULTS HEAV decreased the viability of MCF-7, SKBR-3 and K562 cells (P<0.05). The viability of HEAV-treated K562 cells was reduced compared to HEAV-exposed fibroblasts (P<0.05). Treatment of K562 cells with HEAV induced cell death primarily by late apoptosis and necrosis in assays using annexin V-FITC/7-AAD (P<0.05). The use of Nec-1 and Fer-1 increased the viability of K562 cells treated with HEAV relative to cells exposed to HEAV alone (P<0.01). HEAV-induced Ca2+ release mainly from lysosomes in K562 cells (P<0.01). Furthermore, BAPTA-AM, an intracellular Ca2+ chelator, decreased the number of non-viable cells treated with HEAV (P<0.05). CONCLUSIONS HEAV is cytotoxic and activates several modalities of cell death, which are partially dependent on lysosomal release of Ca2+. These effects may be related to artemisinin and caffeoylquinic acids, the main compounds identified in HEAV.
Collapse
Affiliation(s)
- Lucas Dos Santos Zamarioli
- Department of Pharmacology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP, 04044-020, Brazil
| | - Michele Rosana Maia Santos
- Department of Pharmacology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP, 04044-020, Brazil
| | - Adolfo Garcia Erustes
- Department of Pharmacology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP, 04044-020, Brazil
| | - Vanessa Marques Meccatti
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São Paulo, SP, 12231-280, Brazil
| | - Thaís Cristine Pereira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São Paulo, SP, 12231-280, Brazil
| | - Soraya S Smaili
- Department of Pharmacology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP, 04044-020, Brazil
| | - Maria Cristina Marcucci
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São Paulo, SP, 12231-280, Brazil
| | - Carlos Rocha Oliveira
- Research Group on Phytocomplexes and Cell Signaling, School of Health Sciences, Anhembi Morumbi University, São Paulo, SP, 03164-000, Brazil
| | - Gustavo J S Pereira
- Department of Pharmacology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP, 04044-020, Brazil
| | - Claudia Bincoletto
- Department of Pharmacology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP, 04044-020, Brazil.
| |
Collapse
|
6
|
Liu J, Huang Y, Qian T, Chen J, Ding Y, Lai Z, Zhong X, Lai M, Zhang H, Wang Y, Wang H, Peng Y. Exploring the neuroprotective role of artesunate in mouse models of anti-NMDAR encephalitis: insights from molecular mechanisms and transmission electron microscopy. Cell Commun Signal 2024; 22:269. [PMID: 38745240 PMCID: PMC11094908 DOI: 10.1186/s12964-024-01652-4] [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: 02/20/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The pathway involving PTEN-induced putative kinase 1 (PINK1) and PARKIN plays a crucial role in mitophagy, a process activated by artesunate (ART). We propose that patients with anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis exhibit insufficient mitophagy, and ART enhances mitophagy via the PINK1/PARKIN pathway, thereby providing neuroprotection. METHODS Adult female mice aged 8-10 weeks were selected to create a passive transfer model of anti-NMDAR encephalitis. We conducted behavioral tests on these mice within a set timeframe. Techniques such as immunohistochemistry, immunofluorescence, and western blotting were employed to assess markers including PINK1, PARKIN, LC3B, p62, caspase3, and cleaved caspase3. The TUNEL assay was utilized to detect neuronal apoptosis, while transmission electron microscopy (TEM) was used to examine mitochondrial autophagosomes. Primary hippocampal neurons were cultured, treated, and then analyzed through immunofluorescence for mtDNA, mtROS, TMRM. RESULTS In comparison to the control group, mitophagy levels in the experimental group were not significantly altered, yet there was a notable increase in apoptotic neurons. Furthermore, markers indicative of mitochondrial leakage and damage were found to be elevated in the experimental group compared to the control group, but these markers showed improvement following ART treatment. ART was effective in activating the PINK1/PARKIN pathway, enhancing mitophagy, and diminishing neuronal apoptosis. Behavioral assessments revealed that ART ameliorated symptoms in mice with anti-NMDAR encephalitis in the passive transfer model (PTM). The knockdown of PINK1 led to a reduction in mitophagy levels, and subsequent ART intervention did not alleviate symptoms in the anti-NMDAR encephalitis PTM mice, indicating that ART's therapeutic efficacy is mediated through the activation of the PINK1/PARKIN pathway. CONCLUSIONS At the onset of anti-NMDAR encephalitis, mitochondrial damage is observed; however, this damage is mitigated by the activation of mitophagy via the PINK1/PARKIN pathway. This regulatory feedback mechanism facilitates the removal of damaged mitochondria, prevents neuronal apoptosis, and consequently safeguards neural tissue. ART activates the PINK1/PARKIN pathway to enhance mitophagy, thereby exerting neuroprotective effects and may achieve therapeutic goals in treating anti-NMDAR encephalitis.
Collapse
Affiliation(s)
- Jingsi Liu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yingyi Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Tinglin Qian
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jinyu Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuewen Ding
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhaohui Lai
- Department of Neurology, Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Xinghua Zhong
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Mingjun Lai
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Huili Zhang
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Yuanyuan Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Honghao Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yu Peng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Ma Z, Chen W, Liu Y, Yu L, Mao X, Guo X, Jiang F, Guo Q, Lin N, Zhang Y. Artesunate Sensitizes human hepatocellular carcinoma to sorafenib via exacerbating AFAP1L2-SRC-FUNDC1 axis-dependent mitophagy. Autophagy 2024; 20:541-556. [PMID: 37733919 PMCID: PMC10936616 DOI: 10.1080/15548627.2023.2261758] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 08/25/2023] [Accepted: 09/16/2023] [Indexed: 09/23/2023] Open
Abstract
Sorafenib is the most widely used first-line drug for the treatment of the advanced hepatocellular carcinoma (HCC). Unfortunately, sorafenib resistance often limits its therapeutic efficacy. To evaluate the efficacy of artesunate against sorafenib-resistant HCC and to investigate its underlying pharmacological mechanisms, a "sorafenib resistance related gene-ART candidate target" interaction network was constructed, and a signaling axis consisting with artesunate candidate target AFAP1L2 and sorafenib target SRC, and the downstream FUNDC1-dependent mitophagy was identified as a major contributor to the sorafenib resistance and a potential way of artesunate to mitigate resistance. Notably, our clinical data demonstrated that AFAP1L2 expression in HCC tissues was markedly higher than that in adjacent non-cancerous liver tissues (P < 0.05), and high AFAP1L2 expression was also significantly associated with an unfavorable overall survival of HCC patients (P < 0.05). Experimentally, AFAP1L2 was overexpressed in sorafenib resistant cells, leading to the activation of downstream SRC-FUNDC1 signaling axis, further blocking the FUNDC1 recruitment of LC3B to mitochondria and inhibiting the activation of mitophagy, based on both in vitro and in vivo systems. Moreover, artesunate significantly enhanced the inhibitory effects of sorafenib on resistant cells and tumors by inducing excessive mitophagy. Mechanically, artesunate reduced the expression of AFAP1L2 protein, suppressed the phosphorylation levels of SRC and FUNDC1 proteins, promoted the FUNDC1 recruitment of massive LC3B to mitochondria, and further overactivated the mitophagy and subsequent cell apoptosis of sorafenib resistant cells. In conclusion, artesunate may be a promising strategy to mitigate sorafenib resistance in HCC via exacerbating AFAP1L2-SRC-FUNDC1 axis-dependent mitophagy.Abbreviations: AFAP1L2, actin filament associated protein 1 like 2; ANOVA, analysis of variance; ANXA5, annexin V; ART: artesunate; CETSA, cellular thermal shift assay; CI: combination index; CO-IP: co-immunoprecipitation; CQ: chloroquine; CT, computed tomography; [18F]-FDG, fluoro-2-D-deoxyglucose F18; FUNDC1: FUN14 domain containing 1; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HCC, hepatocellular carcinoma; H&E Staining: hematoxylin - eosin staining; HepG2R, sorafenib resistant HepG2; IF, immunofluorescence; IHC, immunohistochemistry; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; miR, microRNA; mRNA: messenger RNA; OE, overexpression; OS, overall survival; PET, positron emission tomography; qRT-PCR: quantitative real-time PCR; sh, short hairpin; shNC: negative control shRNA; shAFAP1L2: short hairpin AFAP1L2; SORA, sorafenib; SPR, surface plasmon resonance; SRC, SRC proto-oncogene, non-receptor tyrosine kinase; SUV, standardized uptake value; TEM, transmission electron microscopy; TOMM20: translocase of outer mitochondrial membrane 20.
Collapse
Affiliation(s)
- Zhaochen Ma
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjia Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yudong Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lingxiang Yu
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xia Mao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaodong Guo
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Funeng Jiang
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, South China University of Technology, Guangzhou, Guangdong, China
| | - Qiuyan Guo
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Na Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanqiong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Huang Y, Yang Y, Liu G, Xu M. New clinical application prospects of artemisinin and its derivatives: a scoping review. Infect Dis Poverty 2023; 12:115. [PMID: 38072951 PMCID: PMC10712159 DOI: 10.1186/s40249-023-01152-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Recent research has suggested that artemisinin and its derivatives may have therapeutic effects on parasites, viruses, tumors, inflammation and skin diseases. This study aimed to review clinical research on artemisinin and its derivatives except anti-malaria and explore possible priority areas for future development. METHODS Relevant articles in English and Chinese published before 28 October 2021 were reviewed. All articles were retrieved and obtained from databases including WanFang, PubMed/MEDLINE, the Cochrane Library, China National Knowledge International, Embase, OpenGrey, the Grey Literature Report, Grey Horizon, and ClinicalTrials.gov. Studies were selected for final inclusion based on predefined criteria. Information was then extracted and analyzed by region, disease, outcome, and time to identify relevant knowledge gaps. RESULTS Seventy-seven studies on anti-parasitic (35), anti-tumor (16), anti-inflammatory (12), anti-viral (8), and dermatological treatments (7) focused on the safety and efficacy of artemisinin and its derivatives. The anti-parasitic clinical research developed rapidly, with a large number of trials, rapid clinical progress, and multiple research topics. In contrast, anti-viral research was limited and mainly stayed in phase I clinical trials (37.50%). Most of the studies were conducted in Asia (60%), followed by Africa (27%), Europe (8%), and the Americas (5%). Anti-parasite and anti-inflammatory research were mainly distributed in less developed continents such as Asia and Africa, while cutting-edge research such as anti-tumor has attracted more attention in Europe and the United States. At the safety level, 58 articles mentioned the adverse reactions of artemisinin and its derivatives, with only one study showing a Grade 3 adverse event, while the other studies did not show any related adverse reactions or required discontinuation. Most studies have discovered therapeutic effects of artemisinin or its derivatives on anti-parasitic (27), anti-tumor (9), anti-inflammatory (9) and dermatological treatment (6). However, the efficacy of artemisinin-based combination therapies (ACTs) for parasitic diseases (non-malaria) is still controversial. CONCLUSIONS Recent clinical studies suggest that artemisinin and its derivatives may be safe and effective candidates for anti-tumor, anti-parasitic, anti-inflammatory and dermatological drugs. More phase II/III clinical trials of artemisinin and its derivatives on antiviral effects are needed.
Collapse
Affiliation(s)
- Yangmu Huang
- School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China.
- Institute for Global Health and Development, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China.
| | - Yang Yang
- School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
| | - Guangqi Liu
- Energy Saving and Environmental Protection and Occupational Safety and Health Research Institute, China Academy of Railway Sciences Co., Ltd, No. 2 Daliushu Road, Beijing, 100081, China
| | - Ming Xu
- School of Public Health, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, 38 Xue Yuan Road, Haidian District, Beijing, 100191, China
| |
Collapse
|
9
|
Zhang J, Zhou K, Lin J, Yao X, Ju D, Zeng X, Pang Z, Yang W. Ferroptosis-enhanced chemotherapy for triple-negative breast cancer with magnetic composite nanoparticles. Biomaterials 2023; 303:122395. [PMID: 37988899 DOI: 10.1016/j.biomaterials.2023.122395] [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: 06/09/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Triple-negative breast cancer (TNBC) causes great suffering to patients because of its heterogeneity, poor prognosis, and chemotherapy resistance. Ferroptosis is characterized by iron-dependent oxidative damage by accumulating intracellular lipid peroxides to lethal levels, and plays a vital role in the treatment of TNBC based on its intrinsic characteristics. To identify the relationship between chemotherapy resistance and ferroptosis in TNBC, we analyzed the single cell RNA-sequencing public dataset of GSE205551. It was found that the expression of Gpx4 in DOX-resistant TNBC cells was significantly higher than that in DOX-sensitive TNBC cells. Based on this finding, we hypothesize that inducing ferroptosis by inhibiting the expression of Gpx4 can reduce the resistance of TNBC to DOX and enhance the therapeutic effect of chemotherapy on TNBC. Herein, dihydroartemisinin (DHA)-loaded polyglutamic acid-stabilized Fe3O4 magnetic nanoparticles (Fe3O4-PGA-DHA) was combined with DOX-loaded polyaspartic acid-stabilized Fe3O4 magnetic nanoparticles (Fe3O4-PASP-DOX) for ferroptosis-enhanced chemotherapy of TNBC. Compared with Fe3O4-PASP-DOX, Fe3O4-PGA-DHA + Fe3O4-PASP-DOX demonstrated significantly stronger cytotoxicity against different TNBC cell lines and achieved significantly more intracellular accumulation of reactive oxygen species and lipid peroxides. Furthermore, transcriptomic analyses demonstrated that Fe3O4-PASP-DOX-induced apoptosis could be enhanced by Fe3O4-PGA-DHA-induced ferroptosis and Fe3O4-PGA-DHA + Fe3O4-PASP-DOX might trigger ferroptosis in MDA-MB-231 cells by inhibiting the PI3K/AKT/mTOR/GPX4 pathway. Fe3O4-PGA-DHA + Fe3O4-PASP-DOX showed superior anti-tumor efficacy on MDA-MB-231 tumor-bearing mice, providing great potential for improving the therapeutic effect of TNBC.
Collapse
Affiliation(s)
- Jiaxin Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Kaicheng Zhou
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jingbo Lin
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Xianxian Yao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Dianwen Ju
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xian Zeng
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
10
|
Nabi N, Singh S, Saffeullah P. An updated review on distribution, biosynthesis and pharmacological effects of artemisinin: A wonder drug. PHYTOCHEMISTRY 2023; 214:113798. [PMID: 37517615 DOI: 10.1016/j.phytochem.2023.113798] [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: 02/12/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
Plant-based drugs have been used for centuries for treating different ailments. Malaria, one of the prevalent threats in many parts of the world, is treated mainly by artemisinin-based drugs derived from plants of genus Artemisia. However, the distribution of artemisinin is restricted to a few species of the genus; besides, its yield depends on ontogeny and the plant's geographical location. Here, we review the studies focusing on biosynthesis and distributional pattern of artemisinin production in species of the genus Artemisia. We also discussed various agronomic and in vitro methods and molecular approaches to increase the yield of artemisinin. We have summarized different mechanisms of artemisinin involved in its anti-malarial, anti-cancer, anti-inflammatory and anti-viral activities (like against Covid-19). Overall the current review provides a synopsis of a global view of the distribution of artemisinin, its biosynthesis, and pharmacological potential in treating various diseases like malaria, cancer, and coronavirus, which may provoke future research efforts in drug development. Nevertheless, long-term trials and molecular approaches, like CRISPR-Cas, are required for in-depth research.
Collapse
Affiliation(s)
- Neelofer Nabi
- Department of Botany, University of Kashmir, Srinagar, Jammu and Kashmir, 190006, India
| | - Seema Singh
- Department of Botany, University of Kashmir, Srinagar, Jammu and Kashmir, 190006, India
| | - Peer Saffeullah
- Department of Botany, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
11
|
Afshari H, Noori S, Zarghi A. Curcumin potentiates the anti-inflammatory effects of Tehranolide by modulating the STAT3/NF-κB signaling pathway in breast and ovarian cancer cell lines. Inflammopharmacology 2023; 31:2541-2555. [PMID: 37452228 DOI: 10.1007/s10787-023-01281-2] [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: 01/17/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Studies have demonstrated that natural products, such as curcumin and artemisinin, possess anti-inflammatory effects, which can be beneficial for cancer treatment. Tehranolide, as a novel natural product, has a wide range of biological activities, including anti-cancer effects. However, many properties of Tehranolide, like its anti-inflammatory activity and its combination with curcumin, have not been investigated yet. This investigation examined the anti-inflammatory activity of Tehranolide, either alone or in combination with curcumin, via modulating the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and STAT3 (signal transducer and activator of transcription 3) signaling pathways in MDA-MB-231 and SKOV3, breast and ovarian cancer cell lines. METHODS ELISA-based methods were employed to measure the pro-inflammatory cytokine levels and the NF-κB activity in lipopolysaccharide (LPS)-induced cells. The real-time PCR experiment and Griess test were performed to evaluate inducible nitric oxide synthase (iNOS) gene expression and nitrite levels, respectively. The STAT3 and NF-κB signaling pathways were investigated by Western blotting analysis. Tehranolide's anti-cancer activity was also assessed in a mouse model of breast cancer using the TUNEL (terminal deoxynucleotidyl transferase nick-end labeling) assay. RESULTS Tehranolide diminished levels of pro-inflammatory cytokines in cancer cells. Additionally, it suppressed NF-κB DNA binding and STAT3 phosphorylation, reducing iNOS gene expression and nitrite production. Moreover, Western blotting showed that Tehranolide enhanced the inhibitory κB (IκBα) and Bcl-2 (B-cell lymphoma 2)-associated X (BAX) expression, and downregulated the expression of Bcl-2 proteins. Furthermore, the TUNEL assay demonstrated that Tehranolide induced apoptosis in a breast cancer mouse model. Curcumin potentiated all the anti-inflammatory effects of Tehranolide. CONCLUSION This investigation indicated for the first time that Tehranolide, either alone or in combination with curcumin, exerted its anti-inflammatory effects by suppressing NF-κB and STAT3 signaling pathways in SKOV3 and MDA-MB-231 cells.
Collapse
Affiliation(s)
- Havva Afshari
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shokoofe Noori
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Zarghi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Zhang X, Liu H, Li N, Li J, Wang M, Ren X. A (Traditional Chinese Medicine) TCM-Inspired Doxorubicin Resistance Reversing Strategy: Preparation, Characterization, and Application of a Co-loaded pH-Sensitive Liposome. AAPS PharmSciTech 2023; 24:181. [PMID: 37697172 DOI: 10.1208/s12249-023-02630-8] [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: 05/12/2023] [Accepted: 07/25/2023] [Indexed: 09/13/2023] Open
Abstract
In this study, nano-strategy for combined medication of active compounds from traditional Chinese medicine herbs was proposed to achieve the synergistic effects of inhibiting the doxorubicin (DOX) resistance, reducing the cardio-toxicity, and improving the treatment efficacy simultaneously. Dihydroartemisinin (DHA) and tetrandrine (TET) were co-delivered for the first time to treat DOX resistance of breast cancer with multi-pathway mechanism. Tumor micro-environment sensitivity prescription was adopted to enhance the reversal effect of DOX resistance nearly 50 times (resistance index, RI was 46.70) and uptake ability. The DHA-TET pH-sensitive liposomes (DHA-TET pH-sensitive LPs) had a good spherical structure and a uniform dispersion structure with particle size, polydispersity index (PDI), and zeta potential of 112.20 ± 4.80 nm, 0.20 ± 0.02, and - 8.63 ± 0.74 Mv, and was stable until 14 days under the storage environment of 4°C and for 6 months at room temperature environment. With the DOX resistance reversing ability increased, the inhibition effect of DHA-TET pH-sensitive LPs on both MCF-7/ADR cells and MCF-7 cells was significantly enhanced; meanwhile, the toxicity on cardiac cell (H9c2) was lowered. Ferroptosis induced by the DHA was investigated showing that the intracellular reactive oxygen species (ROS) and lipid peroxidation were increased to promote the synergistic effect through the due-loaded nano-carrier, providing a promising alternative for future clinical application.
Collapse
Affiliation(s)
- Xueyan Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hua Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Na Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jiayang Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Meng Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, People's Republic of China.
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
13
|
Ji T, Chen M, Liu Y, Jiang H, Li N, He X. Artesunate alleviates intestinal ischemia/reperfusion induced acute lung injury via up-regulating AKT and HO-1 signal pathway in mice. Int Immunopharmacol 2023; 122:110571. [PMID: 37441813 DOI: 10.1016/j.intimp.2023.110571] [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: 04/12/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023]
Abstract
Acute Lung injury (ALI) is a common complication following intestinal ischemia/reperfusion (II/R) injury that can lead to acute respiratory distress syndrome (ARDS) a fatal illness for there is no specific therapy. The semisynthetic artemisinin Artesunate (Art) extracted from Artemisia annua has been found lots of pharmaceutical effects such as anti-malaria, anti-inflammatory, and anti-apoptosis. This study aimed to investigate the effect of Artesunate on intestinal ischemia/reperfusion and the mechanism of how Artesunate works in mice. To establish the II/R model, the C57BL/c mice were subjected to occlude superior mesenteric artery (SMA) for 45 min and 120 min reperfusion, and the lung tissue was collected for examination. Severe lung injury occurred during the II/R, meanwhile Art pretreatment decreased the lung injury score, wet/dry ratio, the level of MDA, MPO, IL-1β, TNFα, CXCL1, MCP-1, the TUNEL-positive cells, Bax and Cleaved-Caspase3 protein expression obviously, and increased the activity of SOD and the expression of Bcl-2. In addition, the protein of P-AKT and HO-1 were upregulated during the Art pretreatment. Then the AKT inhibitor Triciribin and HO-1 inhibitor Tin-protoporphyrin IX were administered which reversed the protein expression of apoptosis, AKT and HO-1. Our study suggests that Art mitigated the II/R induced acute lung injury by targeting the oxidative stress, inflammatory response and apoptosis which is associated with the activating of AKT and HO-1, providing novel insights into the therapeutic candidate for the treatment of II/R induced acute lung injury.
Collapse
Affiliation(s)
- Tuo Ji
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 169 East Lake Road, Wuhan, Hubei 430071, China; Department of Anesthesiology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Meng Chen
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, 745 Wuluo Road, Wuhan, Hubei 430070, China.
| | - Yinyin Liu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 169 East Lake Road, Wuhan, Hubei 430071, China.
| | - Haixing Jiang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 169 East Lake Road, Wuhan, Hubei 430071, China.
| | - Na Li
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, 745 Wuluo Road, Wuhan, Hubei 430070, China.
| | - Xianghu He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 169 East Lake Road, Wuhan, Hubei 430071, China.
| |
Collapse
|
14
|
Shao X, Meng C, Song W, Zhang T, Chen Q. Subcellular visualization: Organelle-specific targeted drug delivery and discovery. Adv Drug Deliv Rev 2023; 199:114977. [PMID: 37391014 DOI: 10.1016/j.addr.2023.114977] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Organelles perform critical biological functions due to their distinct molecular composition and internal environment. Disorders in organelles or their interacting networks have been linked to the incidence of numerous diseases, and the research of pharmacological actions at the organelle level has sparked pharmacists' interest. Currently, cell imaging has evolved into a critical tool for drug delivery, drug discovery, and pharmacological research. The introduction of advanced imaging techniques in recent years has provided researchers with richer biological information for viewing and studying the ultrastructure of organelles, protein interactions, and gene transcription activities, leading to the design and delivery of precision-targeted drugs. Therefore, this reviews the research on organelles-targeted drugs based upon imaging technologies and development of fluorescent molecules for medicinal purposes. We also give a thorough analysis of a number of subcellular-level elements of drug development, including subcellular research instruments and methods, organelle biological event investigation, subcellular target and drug identification, and design of subcellular delivery systems. This review will make it possible to promote drug research from the individual/cellular level to the subcellular level, as well as give a new focus based on newly found organelle activities.
Collapse
Affiliation(s)
- Xintian Shao
- School of Life Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Caicai Meng
- School of Life Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Wenjing Song
- School of Life Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China; School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Tao Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province 250014, PR China
| | - Qixin Chen
- School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China.
| |
Collapse
|
15
|
Ornelas C, Astruc D. Ferrocene-Based Drugs, Delivery Nanomaterials and Fenton Mechanism: State of the Art, Recent Developments and Prospects. Pharmaceutics 2023; 15:2044. [PMID: 37631259 PMCID: PMC10458437 DOI: 10.3390/pharmaceutics15082044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Ferrocene has been the most used organometallic moiety introduced in organic and bioinorganic drugs to cure cancers and various other diseases. Following several pioneering studies, two real breakthroughs occurred in 1996 and 1997. In 1996, Jaouen et al. reported ferrocifens, ferrocene analogs of tamoxifen, the chemotherapeutic for hormone-dependent breast cancer. Several ferrocifens are now in preclinical evaluation. Independently, in 1997, ferroquine, an analog of the antimalarial drug chloroquine upon the introduction of a ferrocenyl substituent in the carbon chain, was reported by the Biot-Brocard group and found to be active against both chloroquine-sensitive and chloroquine-resistant strains of Plasmodium falciparum. Ferroquine, in combination with artefenomel, completed phase IIb clinical evaluation in 2019. More than 1000 studies have been published on ferrocenyl-containing pharmacophores against infectious diseases, including parasitic, bacterial, fungal, and viral infections, but the relationship between structure and biological activity has been scarcely demonstrated, unlike for ferrocifens and ferroquines. In a majority of ferrocene-containing drugs, however, the production of reactive oxygen species (ROS), in particular the OH. radical, produced by Fenton catalysis, plays a key role and is scrutinized in this mini-review, together with the supramolecular approach utilizing drug delivery nanosystems, such as micelles, metal-organic frameworks (MOFs), polymers, and dendrimers.
Collapse
Affiliation(s)
- Catia Ornelas
- ChemistryX, R&D Department, R&D and Consulting Company, 9000-160 Funchal, Portugal
| | - Didier Astruc
- University of Bordeaux, ISM, UMR CNRS, No. 5255, 351 Cours de la Libération, CEDEX, 33405 Talence, France
| |
Collapse
|
16
|
Xu Q, Duan YY, Pan M, Jin QW, Tao JP, Huang SY. In Vitro Evaluation Reveals Effect and Mechanism of Artemether against Toxoplasma gondii. Metabolites 2023; 13:metabo13040476. [PMID: 37110135 PMCID: PMC10145583 DOI: 10.3390/metabo13040476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Due to the limited effectiveness of existing drugs for the treatment of toxoplasmosis, there is a dire need for the discovery of new therapeutic options. Artemether is an important drug for malaria and several studies have indicated that it also exhibits anti-T. gondii activity. However, its specific effect and mechanisms are still not clear. To elucidate its specific role and potential mechanism, we first evaluated its cytotoxicity and anti-Toxoplasma effect on human foreskin fibroblast cells, and then analyzed its inhibitory activity during T. gondii invasion and intracellular proliferation. Finally, we examined its effect on mitochondrial membrane potential and reactive oxygen species (ROS) in T. gondii. The CC50 value of artemether was found to be 866.4 μM, and IC50 was 9.035 μM. It exhibited anti-T. gondii activity and inhibited the growth of T. gondii in a dose-dependent manner. We also found that the inhibition occurred primarily in intracellular proliferation, achieved by reducing the mitochondrial membrane integrity of T. gondii and stimulating ROS production. These findings suggest that the mechanism of artemether against T. gondii is related to a change in the mitochondrial membrane and the increase in ROS production, which may provide a theoretical basis for optimizing artemether derivatives and further improving their anti-Toxoplasma efficacy.
Collapse
Affiliation(s)
- Qiong Xu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Yin-Yan Duan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Ming Pan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Qi-Wang Jin
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Jian-Ping Tao
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Si-Yang Huang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, and Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
17
|
Zhang R, Kang R, Tang D. Ferroptosis in gastrointestinal cancer: From mechanisms to implications. Cancer Lett 2023; 561:216147. [PMID: 36965540 DOI: 10.1016/j.canlet.2023.216147] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
Ferroptosis is a form of regulated cell death that is initiated by excessive lipid peroxidation that results in plasma membrane damage and the release of damage-associated molecular patterns. In recent years, ferroptosis has gained significant attention in cancer research due to its unique mechanism compared to other forms of regulated cell death, especially caspase-dependent apoptotic cell death. Gastrointestinal (GI) cancer encompasses malignancies that arise in the digestive tract, including the stomach, intestines, pancreas, colon, liver, rectum, anus, and biliary system. These cancers are a global health concern, with high incidence and mortality rates. Despite advances in medical treatments, drug resistance caused by defects in apoptotic pathways remains a persistent challenge in the management of GI cancer. Hence, exploring the role of ferroptosis in GI cancers may lead to more efficacious treatment strategies. In this review, we provide a comprehensive overview of the core mechanism of ferroptosis and discuss its function, regulation, and implications in the context of GI cancers.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
18
|
Zhang M, Lin J, Zhang J, Zhao R, Wan J, Nong Y. Artesunate inhibits airway remodeling in asthma via the MAPK signaling pathway. Front Pharmacol 2023; 14:1145188. [PMID: 36998616 PMCID: PMC10043319 DOI: 10.3389/fphar.2023.1145188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Background: Artesunate (ART), is a semi-synthetic water-soluble artemisinin derivative extracted from the plant Artemisia annua, which is often used to treating malaria. In vivo and in vitro studies suggested it may help decrease inflammation and attenuate airway remodeling in asthma. However, its underlying mechanism of action is not elucidated yet. Herein, an attempt is made to investigate the ART molecular mechanism in treating asthma.Methods: The BALB/c female mice sensitized via ovalbumin (OVA) have been utilized to establish the asthma model, followed by carrying out ART interventions. Lung inflammation scores by Haematoxylin and Eosin (H&E), goblet cell hyperplasia grade by Periodic Acid-Schiff (PAS), and collagen fibers deposition by Masson trichrome staining have been utilized for evaluating how ART affected asthma. RNA-sequencing (RNA-seq) analyses were performed to identify differentially expressed genes (DEGs). The DEGs were analyzed by Gene Ontology (GO) terms, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, and Protein-Protein interaction (PPI) function analyses. Hub clusters were found by Cytoscape MCODE. Subsequently, Real-Time quantitative PCR (RT-qPCR) verified the mRNA expression profiles of DEGs. Finally, immunohistochemistry (IHC) and western blots have validated the relevant genes and potential pathways.Results: ART considerably attenuated inflammatory cell infiltration, mucus secretion, and collagen fibers deposition. KEGG pathway analysis revealed that the ART played a protective role via various pathways including the mitogen-activated protein kinase (MAPK) pathway as one of them. Moreover, ART could alleviate the overexpression of found in inflammatory zone 1(FIZZ1) as revealed by IHC and Western blot analyses. ART attenuated OVA-induced asthma by downregulating phosphorylated p38 MAPK.Conclusion: ART exerted a protective function in a multitarget and multi-pathway on asthma. FIZZ1 was a possible target for asthma airway remodeling. The MARK pathway was one of the key pathways by which ART protected against asthma.
Collapse
Affiliation(s)
- Mengyuan Zhang
- Department of Respiratory and Critical Care, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Jiangtao Lin
- Department of Respiratory and Critical Care, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Jiangtao Lin,
| | - Jingyuan Zhang
- Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ruiheng Zhao
- Department of Respiratory and Critical Care, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Jingxuan Wan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Beijing, China
| | - Ying Nong
- Department of Respiratory and Critical Care, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
19
|
Huang S, Galaj E, Wang J, Guo Y, Wang S, Shi M, Yin X, Liu K, Luo Y, Meng L, Shi H. Repurposing antimalarial artesunate for the prophylactic treatment of depression: Evidence from preclinical research. Brain Behav 2023; 13:e2833. [PMID: 36573693 PMCID: PMC9847619 DOI: 10.1002/brb3.2833] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Several studies have linked inflammation and oxidative stress with the pathogenesis of depression. Artesunate is a commonly used medication to treat malaria and has been shown to produce antioxidant, anti-inflammatory, and immunomodulatory effects. However, its prophylactic effects on depression and depression-related brain pathology are unknown. METHODS In Experiment 1, using a PC12 cell line, we investigated whether artesunate can prevent hydrogen peroxide (H2 O2 )-induced oxidative injury that mimics oxidative stress commonly observed in the depressed brain. Next, using lipopolysaccharide (LPS)-induced mouse model of depression, we investigated whether artesunate can prevent behavioral deficits observed in the open field test, novelty-suppressed feeding test, sucrose preference test, forced swimming test, and tail suspension procedure. RESULTS We found that artesunate significantly prevented a H2 O2 -induced reduction in PC12 cell activity, suggesting its antioxidant potential. We also found that mice pretreated with artesunate (5, 15 mg/kg) intraperitoneally (i.p.) prior to the LPS (.8 mg/kg, i.p.) treatment showed fewer and less severe depression- and anxiety-like behaviors than the LPS-treated control mice. CONCLUSION Our findings indicate that artesunate produces antioxidant effect, as well as antidepressant and anxiolytic effects. Importantly, our findings first demonstrate that artesunate can prevent LPS-induced depression- and anxiety-like symptoms, strongly suggesting its prophylactic potential in the treatment of depression and, perhaps, other psychiatric disorders associated with inflammation and oxidative stress.
Collapse
Affiliation(s)
- Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Ewa Galaj
- Department of Psychological and Brain Sciences, Colgate University, Hamilton, New York, USA
| | - Jinfeng Wang
- Department of Obstetrics and Gynecology, The No.1 Hospital of Yongnian District Handan City, Handan, China
| | - Yi Guo
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China.,Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, China
| | - Shuang Wang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China.,Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, China
| | - Mengxu Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China
| | - Xueyong Yin
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China.,Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, China
| | - Keyao Liu
- Department of Biomedical Engineering, University of North Carolina Chapel Hill, North Carolina, USA
| | - Yixiao Luo
- Hunan Province People's Hospital, The First-affiliated Hospital of Hunan Normal University, Changsha, China
| | - Li Meng
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China.,Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, China
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China.,Hebei Key laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang, China
| |
Collapse
|
20
|
Cheikh IA, El-Baba C, Youssef A, Saliba NA, Ghantous A, Darwiche N. Lessons learned from the discovery and development of the sesquiterpene lactones in cancer therapy and prevention. Expert Opin Drug Discov 2022; 17:1377-1405. [PMID: 36373806 DOI: 10.1080/17460441.2023.2147920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/06/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Sesquiterpene lactones (SLs) are one of the most diverse bioactive secondary metabolites found in plants and exhibit a broad range of therapeutic properties . SLs have been showing promising potential in cancer clinical trials, and the molecular mechanisms underlying their anticancer potential are being uncovered. Recent evidence also points to a potential utility of SLs in cancer prevention. AREAS COVERED This work evaluates SLs with promising anticancer potential based on cell, animal, and clinical models: Artemisinin, micheliolide, thapsigargin dehydrocostuslactone, arglabin, parthenolide, costunolide, deoxyelephantopin, alantolactone, isoalantolactone, atractylenolide 1, and xanthatin as well as their synthetic derivatives. We highlight actionable molecular targets and biological mechanisms underlying the anticancer therapeutic properties of SLs. This is complemented by a unique assessment of SL mechanisms of action that can be exploited in cancer prevention. We also provide insights into structure-activity and pharmacokinetic properties of SLs and their potential use in combination therapies. EXPERT OPINION We extract seven major lessons learned and present evidence-based solutions that can circumvent some scientific limitations or logistic impediments in SL anticancer research. SLs continue to be at the forefront of cancer drug discovery and are worth a joint interdisciplinary effort in order to leverage their potential in cancer therapy and prevention.
Collapse
Affiliation(s)
- Israa A Cheikh
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Chirine El-Baba
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Ali Youssef
- Department of Chemistry, American University of Beirut, Beirut, Lebanon
| | - Najat A Saliba
- Department of Chemistry, American University of Beirut, Beirut, Lebanon
| | - Akram Ghantous
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
21
|
Hua L, Liang S, Zhou Y, Wu X, Cai H, Liu Z, Ou Y, Chen Y, Chen X, Yan Y, Wu D, Sun P, Hu W, Yang Z. Artemisinin-derived artemisitene blocks ROS-mediated NLRP3 inflammasome and alleviates ulcerative colitis. Int Immunopharmacol 2022; 113:109431. [DOI: 10.1016/j.intimp.2022.109431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/23/2022] [Accepted: 11/03/2022] [Indexed: 11/15/2022]
|
22
|
Wu CZ, Gao MJ, Chen J, Sun XL, Zhang KY, Dai YQ, Ma T, Li HM, Zhang YX. Isobavachalcone Induces Multiple Cell Death in Human Triple-Negative Breast Cancer MDA-MB-231 Cells. Molecules 2022; 27:6787. [PMID: 36296386 PMCID: PMC9612085 DOI: 10.3390/molecules27206787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 10/29/2023] Open
Abstract
Standardized treatment guidelines and effective drugs are not available for human triple-negative breast cancer (TNBC). Many efforts have recently been exerted to investigate the efficacy of natural compounds as anticancer agents owing to their low toxicity. However, no study has examined the effects of isobavachalcone (IBC) on the programmed cell death (PCD) of human triple-negative breast MDA-MB-231 cancer cells. In this study, IBC substantially inhibited the proliferation of MDA-MB-231 cells in concentration- and time-dependent manners. In addition, we found that IBC induced multiple cell death processes, such as apoptosis, necroptosis, and autophagy in MDA-MB-231 cells. The initial mechanism of IBC-mediated cell death in MDA-MB-231 cells involves the downregulation of Akt and p-Akt-473, an increase in the Bax/Bcl-2 ratio, and cleaved caspases-3 induced apoptosis; the upregulation of RIP3, p-RIP3 and MLKL induced necroptosis; as well as a simultaneous increase in LC3-II/I ratio induced autophagy. In addition, we observed that IBC induced mitochondrial dysfunction, thereby decreasing cellular ATP levels and increasing reactive oxygen species accumulation to induce PCD. These results suggest that IBC is a promising lead compound with anti-TNBC activity.
Collapse
Affiliation(s)
- Cheng-Zhu Wu
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu 233030, China
| | - Mei-Jia Gao
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
| | - Jie Chen
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
| | - Xiao-Long Sun
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
| | - Ke-Yi Zhang
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
| | - Yi-Qun Dai
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
| | - Tao Ma
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
| | - Hong-Mei Li
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu 233030, China
| | - Yu-Xin Zhang
- Anhui Province Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu 233030, China
- School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China
| |
Collapse
|
23
|
Cong T, Luo Y, Fu Y, Liu Y, Li Y, Li X. New perspectives on ferroptosis and its role in hepatocellular carcinoma. Chin Med J (Engl) 2022; 135:2157-2166. [PMID: 36525603 PMCID: PMC9771279 DOI: 10.1097/cm9.0000000000002327] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Indexed: 12/23/2022] Open
Abstract
ABSTRACT For a long time, the morbidity and mortality rates of hepatocellular carcinoma (HCC) have remained high. Since the concept of ferroptosis was introduced in 2012, researchers' perspectives have shifted toward finding novel ferroptosis-related treatment strategies, especially for tumors that are resistant to apoptosis. In recent years, there have been an increasing number of studies on ferroptosis, and these studies have found that ferroptosis has great potential and promise for cancer treatment. Ferroptosis is a kind of regulated cell death (RCD); unlike apoptosis, ferroptosis is an iron-dependent type of RCD driven by lipid peroxidation. The whole process of ferroptosis mainly revolves around three pathways (system xc-/ glutathione peroxidase 4 [GPX4]), lipid peroxidation, and iron metabolism), which are also regulated by various metabolic factors. This review will attempt to analyze the relationship between the system xc-/GPX4 pathway, lipid peroxidation, iron metabolism, and ferroptosis from three aspects (triggering, execution, and regulation), and the regulatory factors for ferroptosis will be summarized. In this review, we will also illustrate the relationship between ferroptosis and tumors as well as its application in tumors from the perspective of HCC. Finally, we will summarize the current limitations and needs and provide perspectives related to the focus of development in the future.
Collapse
Affiliation(s)
- Tianhao Cong
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yingen Luo
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan Fu
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Liu
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yujie Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
24
|
Liu T, Jin Q, Ren F, Yang L, Mao H, Ma F, Wang Y, Li P, Zhan Y. Potential therapeutic effects of natural compounds targeting autophagy to alleviate podocyte injury in glomerular diseases. Biomed Pharmacother 2022; 155:113670. [PMID: 36116248 DOI: 10.1016/j.biopha.2022.113670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/02/2022] Open
Abstract
Podocyte injury is a common cause of proteinuric kidney diseases. Uncontrollable progressive podocyte loss accelerates glomerulosclerosis and increases the risk of end-stage renal disease. To date, owing to the complex pathological mechanism, effective therapies for podocyte injury have been limited. Accumulating evidence supports the indispensable role of autophagy in the maintenance of podocyte homeostasis. A variety of natural compounds and their derivatives have been found to regulate autophagy through multiple targets, including promotes nuclear transfer of transcription factor EB and lysosomal repair. Here, we reviewed the recent studies on the use of natural compounds and their derivatives as autophagy regulators and discussed their potential applications in ameliorating podocyte injury. Several known natural compounds with autophagy-regulatory properties, such as quercetin, silibinin, kaempferol, and artemisinin, and their medical uses were also discussed. This review will help in improving the understanding of the podocyte protective mechanism of natural compounds and promote their development for clinical use.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feihong Ren
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China.
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
25
|
张 玉, 高 美, 朱 美, 李 红, 马 涛, 吴 成. [Isobavachalcone induces cell death through multiple pathways in human breast cancer MCF-7 cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:878-885. [PMID: 35790438 PMCID: PMC9257353 DOI: 10.12122/j.issn.1673-4254.2022.06.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the effects of isobavachalcone (IBC) on cell death of human breast cancer MCF-7 cells and explore the possible mechanism. METHODS MCF-7 cells were treated with different concentrations of IBC, and the changes in cell proliferation were assessed using MTT assay. Apoptosis of MCF-7 cells following treatment with 10, 20, and 40 μmol/L IBC was analyzed using flow cytometry with annexin V-FITC/PI double staining and fluorescence microscopy, and the expressions of apoptosis- and autophagy-related proteins (Bax, Bcl-2, Akt, p-Akt, p62, and LC3) were detected with Western blotting. Electron microscopy was used to observe the changes in submicrostructure of the cells following treatment with 40 μmol/L IBC. JC-1 assay kit, ATP assay kit, and reactive oxygen species (ROS) kit were used to determine the effect of IBC on mitochondrial function of the cells. RESULTS MTT assay showed that IBC significantly inhibited the proliferation of MCF-7 cells in a concentration- and time-dependent manner, with IC50 values of 38.46, 31.31, and 28.26 μmol/L at 24, 48, and 72 h, respectively. IBC also concentration-dependently induced apoptosis of MCF-7 cells. IBC-induced cell death was inhibited by z-VAD-fmk, a caspase inhibitor (P < 0.05), but not by the necroptosis inhibitor necrostatin-1 (Nec-1). Western blotting showed that IBC-induced MCF-7 cell apoptosis by increasing Bax expression and down-regulating the expressions of Bcl-2, Akt and p-Akt-473 (all P < 0.05). With the increase of IBC concentration, the expression of autophagy-related protein p62 and the LC3-II/I ratio increased progressively. Electron microscopy revealed the presence of autophagic bodies in IBC-treated MCF-7 cells. IBC treatment also resulted in decreased mitochondrial membrane potential and intracellular ATP level and increased ROS accumulation in MCF-7 cells (P < 0.05). CONCLUSION IBC is capable of inducing both apoptosis and autophagy in MCF-7 cells, suggesting the potential value of IBC as a lead compound in the development of anti-breast cancer agents.
Collapse
Affiliation(s)
- 玉心 张
- 蚌埠医学院检验医学院,安徽 蚌埠 233030School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 美佳 高
- 蚌埠医学院药学院,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - 美林 朱
- 蚌埠医学院药学院,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - 红梅 李
- 蚌埠医学院药学院,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - 涛 马
- 蚌埠医学院药学院,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - 成柱 吴
- 蚌埠医学院药学院,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| |
Collapse
|
26
|
Bai S, Zhou J, Nong X, Shi R, Yuan Z, Ma C, Li J. Mechanism and effects of artesunate on the liver function of rats with type 1 diabetic periodontitis. Can J Physiol Pharmacol 2022; 100:741-754. [PMID: 35500287 DOI: 10.1139/cjpp-2021-0665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Periodontitis is an inflammatory disease of the gums. Periodontitis in patients with diabetes can aggravate insulin resistance, but its molecular and biological mechanism remains unclear. This study aimed to explore the effects of diabetic periodontitis on liver function and determine the mechanism by which artesunate improves liver function. Rats with streptozotocin-induced diabetes were divided into five groups, i.e., normal control group (NC group), diabetic periodontitis group (DM+PD group), artesunate intervention group (ART group), insulin intervention group (INS group), and combined medication intervention group (ART+INS group). Drug interventions were then administered to the rats in each group as follows: 50 mg/kg artesunate to the ART group, 6 U/kg insulin to the INS group, and 50 mg/kg artesunate + 6 U/kg insulin to the ART+INS group. Blood samples, liver tissues, and the maxillary alveolar bone were collected post-sacrifice. ART was found to significantly ameliorate hyperglycemia, blood lipid levels, and liver function. The levels of inflammatory factors reduced; the effect was more pronounced in the ART+INS group. Artesunate presumably inhibits the TLR4/NF-κB signaling pathway and expression of downstream inflammatory factors, thereby exerting a protective effect on diabetes-related liver function. This offers a fresh approach to treat diabetes mellitus.
Collapse
Affiliation(s)
- Shuoqiu Bai
- Guangxi Medical University, 74626, Nanning, Guangxi, China;
| | - Jingjing Zhou
- Guangxi Medical University, 74626, Nanning, Guangxi, China;
| | - Xiaolin Nong
- Guangxi Medical University, 74626, Nanning, China;
| | - Rongkang Shi
- Guangxi Medical University, 74626, Nanning, Guangxi, China;
| | - Zhong Yuan
- Guangxi Medical University, 74626, Nanning, Guangxi, China;
| | - Chubin Ma
- Guangxi Medical University, 74626, Nanning, Guangxi, China;
| | - Jiaquan Li
- Guangxi Medical University, 74626, Nanning, Guangxi, China;
| |
Collapse
|
27
|
Feng LS, Cheng JB, Su WQ, Li HZ, Xiao T, Chen DA, Zhang ZL. Cinnamic acid hybrids as anticancer agents: A mini-review. Arch Pharm (Weinheim) 2022; 355:e2200052. [PMID: 35419808 DOI: 10.1002/ardp.202200052] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
Abstract
Cancer, as a long-lasting and dramatic disease, affects almost one-third of human beings globally. Chemotherapeutics play an important role in cancer treatment, but multidrug resistance and severe adverse effects have already become the main causes of failure in tumor chemotherapy. Therefore, it is an urgent need to develop novel chemotherapeutics. Cinnamic acid contains a ubiquitous α,β-unsaturated acid moiety presenting potential therapeutic effects in the treatment of cancer as these derivatives could act on cancer cells by diverse mechanisms of action. Accordingly, cinnamic acid derivatives are critical scaffolds in discovering novel anticancer agents. This review provides a comprehensive overview of cinnamic acid hybrids as anticancer agents. The structure-activity relationship, as well as the mechanisms of action, are also discussed, covering articles published from 2012 to 2021.
Collapse
Affiliation(s)
- Lian-Shun Feng
- WuXi AppTec Co., Ltd., Wuhan, Peoples' Republic of China
| | - Jin-Bo Cheng
- WuXi AppTec Co., Ltd., Wuhan, Peoples' Republic of China
| | - Wen-Qi Su
- WuXi AppTec Co., Ltd., Wuhan, Peoples' Republic of China
| | - Hong-Ze Li
- WuXi AppTec Co., Ltd., Chengdu, Peoples' Republic of China
| | - Tao Xiao
- WuXi AppTec Co., Ltd., Chengdu, Peoples' Republic of China
| | - De-An Chen
- WuXi AppTec Co., Ltd., Wuhan, Peoples' Republic of China
| | - Zhi-Liu Zhang
- WuXi AppTec Co., Ltd., Shanghai, Peoples' Republic of China
| |
Collapse
|
28
|
Shi Q, Xia F, Wang Q, Liao F, Guo Q, Xu C, Wang J. Discovery and repurposing of artemisinin. Front Med 2022; 16:1-9. [PMID: 35290595 PMCID: PMC8922983 DOI: 10.1007/s11684-021-0898-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2022]
Abstract
Malaria is an ancient infectious disease that threatens millions of lives globally even today. The discovery of artemisinin, inspired by traditional Chinese medicine (TCM), has brought in a paradigm shift and been recognized as the “best hope for the treatment of malaria” by World Health Organization. With its high potency and low toxicity, the wide use of artemisinin effectively treats the otherwise drug-resistant parasites and helps many countries, including China, to eventually eradicate malaria. Here, we will first review the initial discovery of artemisinin, an extraordinary journey that was in stark contrast with many drugs in western medicine. We will then discuss how artemisinin and its derivatives could be repurposed to treat cancer, inflammation, immunoregulation-related diseases, and COVID-19. Finally, we will discuss the implications of the “artemisinin story” and how that can better guide the development of TCM today. We believe that artemisinin is just a starting point and TCM will play an even bigger role in healthcare in the 21st century.
Collapse
Affiliation(s)
- Qiaoli Shi
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fei Xia
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qixin Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fulong Liao
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qiuyan Guo
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Chengchao Xu
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China. .,Department of Geriatrics, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, 518020, China.
| | - Jigang Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China. .,Central People's Hospital of Zhanjiang, Zhanjiang, 524045, China. .,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Department of Geriatrics, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, 518020, China.
| |
Collapse
|
29
|
Belardinelli JM, Verma D, Li W, Avanzi C, Wiersma CJ, Williams JT, Johnson BK, Zimmerman M, Whittel N, Angala B, Wang H, Jones V, Dartois V, de Moura VCN, Gonzalez-Juarrero M, Pearce C, Schenkel AR, Malcolm KC, Nick JA, Charman SA, Wells TNC, Podell BK, Vennerstrom JL, Ordway DJ, Abramovitch RB, Jackson M. Therapeutic efficacy of antimalarial drugs targeting DosRS signaling in Mycobacterium abscessus. Sci Transl Med 2022; 14:eabj3860. [PMID: 35196022 DOI: 10.1126/scitranslmed.abj3860] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A search for alternative Mycobacterium abscessus treatments led to our interest in the two-component regulator DosRS, which, in Mycobacterium tuberculosis, is required for the bacterium to establish a state of nonreplicating, drug-tolerant persistence in response to a variety of host stresses. We show here that the genetic disruption of dosRS impairs the adaptation of M. abscessus to hypoxia, resulting in decreased bacterial survival after oxygen depletion, reduced tolerance to a number of antibiotics in vitro and in vivo, and the inhibition of biofilm formation. We determined that three antimalarial drugs or drug candidates, artemisinin, OZ277, and OZ439, can target DosS-mediated hypoxic signaling in M. abscessus and recapitulate the phenotypic effects of genetically disrupting dosS. OZ439 displayed bactericidal activity comparable to standard-of-care antibiotics in chronically infected mice, in addition to potentiating the activity of antibiotics used in combination. The identification of antimalarial drugs as potent inhibitors and adjunct inhibitors of M. abscessus in vivo offers repurposing opportunities that could have an immediate impact in the clinic.
Collapse
Affiliation(s)
- Juan Manuel Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Deepshikha Verma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Wei Li
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Charlotte Avanzi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Crystal J Wiersma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - John T Williams
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | | | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Nicholas Whittel
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Bhanupriya Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Han Wang
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Victoria Jones
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Vinicius C N de Moura
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Camron Pearce
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Alan R Schenkel
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Kenneth C Malcolm
- Department of Medicine, National Jewish Health, Denver, CO, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jerry A Nick
- Department of Medicine, National Jewish Health, Denver, CO, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | | | - Brendan K Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | | | - Diane J Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Robert B Abramovitch
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
30
|
Zhang Y, Wang Y, Li Y, Huang C, Xiao X, Zhong Z, Tang J, Lu H, Tang Y, Yang J. Dihydroartemisinin and Artesunate Inhibit Aerobic Glycolysis via Suppressing c-Myc Signaling in non-small Cell Lung Cancer. Biochem Pharmacol 2022; 198:114941. [PMID: 35149053 DOI: 10.1016/j.bcp.2022.114941] [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: 12/08/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/02/2022]
Abstract
Non-small cell lung cancer (NSCLC) cells exhibit aberrant metabolism characterized by high glycolysis even in the presence of abundant oxygen. Inhibition of aerobic glycolysis remains challenging when identifying potential cancer-specific inhibitors while maintaining or even boosting the anti-cancer immunity. Artemisinin derivatives DHA and AS have shown excellent anti-tumor and immunoenhancing roles in numerous malignancies, but the molecular mechanism of DHA and AS in regulating cancer glucose metabolism is largely unknown. In this study, we proved that DHA and AS inhibit NSCLC growth via prohibiting cancer cell aerobic glycolysis through ERK/c-Myc pathway. First, we proved that DHA and AS have comparable anti-cancer growth roles in both NSCLC cell lines and mouse Lewis Lung Cancer model. Then, our data clearly showed that DHA and AS dose- and time-dependently reduce the uptake of glucose, the production of ATP, and the secretion of lactate, the expression of glucose transporter GLUT1 and two key glycolysis-related enzymes hexokinase and lactate dehydrogenase, as well as the level of c-Myc. Finally, we generated c-Mychigh stable-expressing NSCLC cell line and treated it with DHA or AS, respectively. Our data clearly showed that c-Myc overexpression can partially reverse the glycolysis-repressing role of DHA and AS which strongly supported our proposal that AS and DHA suppress aerobic glycolysis in a c-Myc-dependent manner in NSCLC cells. This study extends our knowledge of artemisinin derivatives in regulating tumor glucose metabolism and provides potential strategy in the therapy of lung cancer.
Collapse
Affiliation(s)
- Yuxi Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cong Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaoqian Xiao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhanqiong Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jingyi Tang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haolan Lu
- School of Medical and Life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yibei Tang
- School of Medical and Life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiahui Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
31
|
Ji P, Wang X, Yin J, Yao Y, Du W. Amplification of Ferroptosis with Liposomal Nanoreactor Cooperates with Low-Toxic Doxorubicin Apoptosis for Enhanced Tumor Chemotherapy. Biomater Sci 2022; 10:1544-1553. [DOI: 10.1039/d2bm00079b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite the remarkable tumor inhibition effect of doxorubicin (DOX), its cardiotoxicity severly limits the clinical dosage and further impairs the chemotherapy efficacy. To improve the biosafety and effectiveness of conventional...
Collapse
|
32
|
Xue P, Chen Q, Ren X, Liu D, Yang X. A novel protoapigenone analog RY10-4 induces apoptosis of breast cancer cells by exacerbating mitochondrial Ca 2+ influx through mitochondrial calcium uniporter. Toxicol Appl Pharmacol 2021; 433:115776. [PMID: 34717963 DOI: 10.1016/j.taap.2021.115776] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/18/2021] [Accepted: 10/24/2021] [Indexed: 12/24/2022]
Abstract
RY10-4, a novel protoapigenone analog with a specific nonaromatic B-ring, displayed enhanced cytotoxicity in various tumor cells, especially for breast cancer cells, but the underlying mechanism remains unclear. In the present study, we confirmed the pro-apoptotic effect of RY10-4 on breast cancer cells. Furthermore, mitochondrial calcium uniporter (MCU) was proved to be up-regulated in RY10-4-treated MDA-MB-231 cells, which resulted in the overload of mitochondrial calcium ([Ca2+]m) and subsequently disrupted mitochondrial functions (characterized by mitochondrial reactive oxygen species (mtROS) accumulation, membrane potential (ΔΨm) depolarization and permeability transition pore (mPTP) opening). And finally, the mitochondrial apoptosis was activated by the release of cytochrome C. Interestingly, knockdown of MCU attenuated the overload of [Ca2+]m and blocked the apoptosis of MDA-MB-231 cells induced by RY10-4, which was consistent with the in vivo results. Taken together, this study proved that RY10-4 could induce apoptosis of breast cancer cells by elevating [Ca2+]m through MCU. Our work contributed previously unknown insights into the mechanisms involving in the clinical efficacy of RY10-4 on breast cancer cells, which also advanced calcium homeostasis as a potential target for chemotherapeutic drugs.
Collapse
Affiliation(s)
- Pingping Xue
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Chen
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiuhua Ren
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dong Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
33
|
Sharma B, Kumar V. Has Ferrocene Really Delivered Its Role in Accentuating the Bioactivity of Organic Scaffolds? J Med Chem 2021; 64:16865-16921. [PMID: 34792350 DOI: 10.1021/acs.jmedchem.1c00390] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Ferrocene is an important structural core in bioorganometallic chemistry because of its inherent stability, excellent redox properties, and low toxicity. Ferroquine and ferrocifen are two of the most notable contributions of ferrocene to medicinal chemistry with remarkable antimalarial and anticancer properties. The improved medicinal properties of these drug candidates highlight the impact that ferrocene can have on the molecular and biological properties of the bioactive compounds. In this Perspective, we investigate the scope and limitations of ferrocene incorporation into organic compounds/natural products on their mode of action and biological activities. We have also discussed the detailed role of ferrocene modifications in influencing the anticancer, antimalarial, and antimicrobial properties of various bioactive moieties to design safer and promising ferrocene-based drugs.
Collapse
Affiliation(s)
- Bharvi Sharma
- Department of Chemistry, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| |
Collapse
|
34
|
Zou X, Liu C, Li C, Fu R, Xu W, Bian H, Dong X, Zhao X, Xu Z, Zhang J, Shen Z. Study on the structure-activity relationship of dihydroartemisinin derivatives: Discovery, synthesis, and biological evaluation of dihydroartemisinin-bile acid conjugates as potential anticancer agents. Eur J Med Chem 2021; 225:113754. [PMID: 34399390 DOI: 10.1016/j.ejmech.2021.113754] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 11/20/2022]
Abstract
A series of dihydroartemisinin derivatives was synthesized, and their anti-proliferation activity against cancer cells was evaluated. Structure-activity relationship studies led to the discovery of dihydroartemisinin-bile acid conjugates that exhibit broad-spectrum anti-proliferation activities. Among them, the dihydroartemisinin-ursodeoxycholic acid conjugate (49) was the most potent, with IC50 values between 0.04 and 0.96 μM when tested to determine its inhibitory properties against 15 various cancer cell lines. In vivo experiments showed that compound 49 effectively suppressed tumor growth in an A549 cell xenograft model at the dosage of 10 mg/kg body weight and in Lewis lung cancer cell transplant model at the dosage of 12 mg/kg body weight.
Collapse
Affiliation(s)
- Xiaosu Zou
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 CaiLun Road, Shanghai, 201203, China
| | - Chang Liu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South WanPing Road, Shanghai, 200032, China
| | - Congcong Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 CaiLun Road, Shanghai, 201203, China
| | - Rong Fu
- School of Medicine, Shanghai Jiao Tong University, 280 South Chongqing Road, Shanghai, 200025, China
| | - Wei Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 CaiLun Road, Shanghai, 201203, China
| | - Hongzhu Bian
- Yunnan Baiyao Group Co. Ltd, 3686 Yunnan Baiyao Street, Kunming, 650200, China
| | - Xun Dong
- Yunnan Baiyao Group Co. Ltd, 3686 Yunnan Baiyao Street, Kunming, 650200, China
| | - Xiaozhen Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South WanPing Road, Shanghai, 200032, China
| | - Zhenye Xu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South WanPing Road, Shanghai, 200032, China.
| | - Jinghua Zhang
- School of Medicine, Shanghai Jiao Tong University, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Zhengwu Shen
- School of Medicine, Shanghai Jiao Tong University, 280 South Chongqing Road, Shanghai, 200025, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 CaiLun Road, Shanghai, 201203, China.
| |
Collapse
|
35
|
The redox mechanism of ferrocene and its phytochemical and biochemical compounds in anticancer therapy: A mini review. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2021.109044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
36
|
Tang J, Zhang X, Cheng L, Liu Y, Chen Y, Jiang Z, Liu J. Multiple stimuli-responsive nanosystem for potent, ROS-amplifying, chemo-sonodynamic antitumor therapy. Bioact Mater 2021; 15:355-371. [PMID: 35356815 PMCID: PMC8935089 DOI: 10.1016/j.bioactmat.2021.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 11/30/2022] Open
Abstract
Although sonodynamic therapy (SDT) is a promising non-invasive tumor treatment strategy due to its safety, tissue penetration depth and low cost, the hypoxic tumor microenvironment limits its therapeutic effects. Herein, we have designed and developed an oxygen-independent, ROS-amplifying chemo-sonodynamic antitumor therapy based on novel pH/GSH/ROS triple-responsive PEG-PPMDT nanoparticles. The formulated artemether (ART)/Fe3O4-loaded PEG-PPMDT NPs can rapidly release drug under the synergistic effect of acidic endoplasmic pH and high intracellular GSH/ROS levels to inhibit cancer cell growth. Besides, the ROS level in the NPs-treated tumor cells is magnified by ART via interactions with both Fe2+ ions formed in situ at acidic pH and external ultrasound irradiation, which is not affected by hypoxia tumor microenvironment. Consequently, the enriched intracellular ROS level can cause direct necrosis of ROS-stressed tumor cells and further accelerate the drug release from the ROS-responsive PEG-PPMDT NPs, achieving an incredible antitumor potency. Specifically, upon the chemo-sonodynamic therapy by ART/Fe3O4-loaded PEG-PPMDT NPs, all xenotransplants of human hepatocellular carcinoma (HepG2) in nude mice shrank significantly, and 40% of the tumors were completely eliminated. Importantly, the Fe3O4 encapsulated in the NPs is an efficient MRI contrast agent and can be used to guide the therapeutic procedures. Further, biosafety analyses show that the PEG-PPMDT NPs possess minimal toxicity to main organs. Thus, our combined chemo-sonodynamic therapeutic method is promising for potent antitumor treatment by controlled release of drug and facile exogenous generation of abundant ROS at target tumor sites. pH/GSH/ROS triple-responsive PEG-PPMDT were synthesized by enzymatic polymerization. ART and Fe3O4 loaded PEG-PPMDT NPs processes SDT/CDT and MRI theranostic function. Intracellular ROS was magnified by Fe2+-ART interaction and ultrasound irradiation.
Collapse
Affiliation(s)
- JunJie Tang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Xiaoge Zhang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Lili Cheng
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Yadong Liu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - You Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Zhaozhong Jiang
- Faculty of Arts and Sciences, Department of Biomedical Engineering, Integrated Science and Technology Center, Yale University, 600 West Campus Drive, West Haven, CT, 06516, United States
- Corresponding author.
| | - Jie Liu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- Corresponding author.
| |
Collapse
|
37
|
Targeting lysosomes in human disease: from basic research to clinical applications. Signal Transduct Target Ther 2021; 6:379. [PMID: 34744168 PMCID: PMC8572923 DOI: 10.1038/s41392-021-00778-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/26/2021] [Indexed: 01/18/2023] Open
Abstract
In recent years, accumulating evidence has elucidated the role of lysosomes in dynamically regulating cellular and organismal homeostasis. Lysosomal changes and dysfunction have been correlated with the development of numerous diseases. In this review, we interpreted the key biological functions of lysosomes in four areas: cellular metabolism, cell proliferation and differentiation, immunity, and cell death. More importantly, we actively sought to determine the characteristic changes and dysfunction of lysosomes in cells affected by these diseases, the causes of these changes and dysfunction, and their significance to the development and treatment of human disease. Furthermore, we outlined currently available targeting strategies: (1) targeting lysosomal acidification; (2) targeting lysosomal cathepsins; (3) targeting lysosomal membrane permeability and integrity; (4) targeting lysosomal calcium signaling; (5) targeting mTOR signaling; and (6) emerging potential targeting strategies. Moreover, we systematically summarized the corresponding drugs and their application in clinical trials. By integrating basic research with clinical findings, we discussed the current opportunities and challenges of targeting lysosomes in human disease.
Collapse
|
38
|
Rein T. Harnessing autophagy to fight SARS-CoV-2: An update in view of recent drug development efforts. J Cell Biochem 2021; 123:155-160. [PMID: 34668225 PMCID: PMC9088732 DOI: 10.1002/jcb.30166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 10/12/2021] [Indexed: 01/18/2023]
Abstract
Drug repurposing is an attractive option for identifying new treatment strategies, in particular in extraordinary situations of urgent need such as the current coronavirus disease 2019 (Covid-19) pandemic. Recently, the World Health Organization announced testing of three drugs as potential Covid-19 therapeutics that are known for their dampening effect on the immune system. Thus, the underlying concept of selecting these drugs is to temper the potentially life-threatening overshooting of the immune system reacting to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. This viewpoint discusses the possibility that the impact of these and other drugs on autophagy contributes to their therapeutic effect by hampering the SARS-CoV-2 life cycle.
Collapse
Affiliation(s)
- Theo Rein
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
39
|
Chen Y, Wang F, Wu P, Gong S, Gao J, Tao H, Shen Q, Wang S, Zhou Z, Jia Y. Artesunate induces apoptosis, autophagy and ferroptosis in diffuse large B cell lymphoma cells by impairing STAT3 signaling. Cell Signal 2021; 88:110167. [PMID: 34628002 DOI: 10.1016/j.cellsig.2021.110167] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/25/2021] [Accepted: 10/05/2021] [Indexed: 02/08/2023]
Abstract
Artesunate (ART), a water-soluble derivative of artemisinin, has been reported to exert antineoplastic effects via diverse mechanisms in various types of cancer. Therefore, understanding the underlying mechanism of action of ART in distinct cancer types is indispensable to optimizing the therapeutic application of ART for different types of cancer. The present study aimed to investigate the cellular and molecular mechanisms responsible for the antineoplastic effects of ART in diffuse large B cell lymphoma (DLBCL) cells. Cell proliferation was measured using Cell Counting Kit-8 and colony formation assays. The levels of apoptosis and cell cycle distribution were investigated using flow cytometry. In addition, western blotting was used to analyze the expression levels of ART-induced apoptosis-, autophagy- and ferroptosis-related proteins. Monodansylcadaverine staining was performed to determine the levels of autophagy. Moreover, malondialdehyde and reactive oxygen species assays were used to determine the levels of ferroptosis. The results of the present study revealed that ART inhibited proliferation, and induced apoptosis, cell cycle arrest, autophagy and ferroptosis in DLBCL cells. Pharmacological inhibition of autophagy and ferroptosis alleviated the increased levels of apoptosis induced by ART. Notably, ART was found to exert its effects via inhibition of STAT3 activation. The genetic knockdown of STAT3 enhanced ART-induced autophagy and ferroptosis, and concomitantly upregulated the expression levels of apoptosis- and cell cycle-related proteins. In conclusion, the findings of the current study suggested that ART may induce apoptosis and cell cycle arrest to inhibit cell proliferation, and regulate autophagy and ferroptosis via impairing the STAT3 signaling pathway in DLBCL cells.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Fujue Wang
- Department of Hematology, The First Affiliated Hospital of University of South China, Hengyang 421001, Hunan, China
| | - Pengqiang Wu
- Department of Hematology, The First Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shuaige Gong
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jie Gao
- Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Huan Tao
- Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Qianqing Shen
- Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuoting Wang
- Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhencang Zhou
- Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yongqian Jia
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
40
|
Zuo Q, Liao L, Yao ZT, Liu YP, Wang DK, Li SJ, Yin XF, He QY, Xu WW. Targeting PP2A with lomitapide suppresses colorectal tumorigenesis through the activation of AMPK/Beclin1-mediated autophagy. Cancer Lett 2021; 521:281-293. [PMID: 34509534 DOI: 10.1016/j.canlet.2021.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/08/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide, and effective therapy remains a challenge. In this study, we take advantage of a drug repurposing strategy to screen small molecules with novel anticancer activities in a small-molecule library consisting of 1056 FDA-approved drugs. We show, for the first time, that lomitapide, a lipid-lowering agent, exhibits antitumor properties in vitro and in vivo. Activated autophagy is characterized as a key biological process in lomitapide-induced CRC repression. Mechanistically, lomitapide stimulated mitochondrial dysfunction-mediated AMPK activation, resulting in increased AMPK phosphorylation and enhanced Beclin1/Atg14/Vps34 interactions, provoking autophagy induction. Autophagy inhibition or AMPK silencing significantly abrogated lomitapide-induced cell death, indicating the significance of AMPK-regulated autophagy in the antitumor activities of lomitapide. More importantly, PP2A was identified as a direct target of lomitapide by limited proteolysis-mass spectrometry (LiP-SMap), and the bioactivity of lomitapide was attenuated in PP2A-deficient cells, suggesting that the anticancer effect of lomitapide occurs in a PP2A-dependent manner. Taken together, the results of the study reveal that lomitapide can be repositioned as a potential therapeutic drug for CRC treatment.
Collapse
Affiliation(s)
- Qian Zuo
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Long Liao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zi-Ting Yao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Ya-Ping Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Ding-Kang Wang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Shu-Jun Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xing-Feng Yin
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wen-Wen Xu
- MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
41
|
Supercritical-derived artemisinin microfibers and microparticles for improving anticancer effects. J Supercrit Fluids 2021. [DOI: 10.1016/j.supflu.2021.105276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Sun X, Hong Y, Shu Y, Wu C, Ye G, Chen H, Zhou H, Gao R, Zhang J. The involvement of Parkin-dependent mitophagy in the anti-cancer activity of Ginsenoside. J Ginseng Res 2021; 46:266-274. [PMID: 35509820 PMCID: PMC9058836 DOI: 10.1016/j.jgr.2021.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 01/03/2023] Open
Abstract
Colon cancer, the third most frequent occurred cancer, has high mortality and extremely poor prognosis. Ginsenoside, the active components of traditional Chinese herbal medicine Panax ginseng, exerts antitumor effect in various cancers, including colon cancer. However, the detailed molecular mechanism of Ginsenoside in the tumor suppression have not been fully elucidated. Here, we chose the representative ginsenoside Rg3 and reported for the first time that Rg3 induces mitophagy in human colon cancer cells, which is responsible for its anticancer effect. Rg3 treatment leads to mitochondria damage and the formation of mitophagosome; when autophagy is inhibited, the clearance of damaged mitochondria can be reversed. Next, our results showed that Rg3 treatment activates the PINK1-Parkin signaling pathway and recruits Parkin and ubiquitin proteins to mitochondria to induce mitophagy. GO analysis of Parkin targets showed that Parkin interacts with a large number of mitochondrial proteins and regulates the molecular function of mitochondria. The cellular energy metabolism enzyme GAPDH is validated as a novel substrate of Parkin, which is ubiquitinated by Parkin. Moreover, GAPDH participates in the Rg3-induced mitophagy and regulates the translocation of Parkin to mitochondria. Functionally, Rg3 exerts the inhibitory effect through regulating the nonglycolytic activity of GAPDH, which could be associated with the cellular oxidative stress. Thus, our results revealed GAPDH ubiquitination by Parkin as a crucial mechanism for mitophagy induction that contributes to the tumor-suppressive function of ginsenoside, which could be a novel treatment strategy for colon cancer.
Collapse
Affiliation(s)
- Xin Sun
- Department of Oncology, Cancer Center of Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | | | - Yuhan Shu
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Caixia Wu
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Guiqin Ye
- Hangzhou Medical College, Hangzhou, China
| | | | - Hongying Zhou
- Department of Oncology, Cancer Center of Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ruilan Gao
- Institution of Hematology Research, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Corresponding author. Institution of Hematology Research, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jianbin Zhang
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- Corresponding author. Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, People’s Hospital of Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
43
|
Huang H, He Q, Guo B, Xu X, Wu Y, Li X. Progress in Redirecting Antiparasitic Drugs for Cancer Treatment. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2747-2767. [PMID: 34188451 PMCID: PMC8235938 DOI: 10.2147/dddt.s308973] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
Drug repurposing is a feasible strategy in developing novel medications. Regarding the cancer field, scientists are continuously making efforts to redirect conventional drugs into cancer treatment. This approach aims at exploring new applications in the existing agents. Antiparasitic medications, including artemisinin derivatives (ARTs), quinine-related compounds, niclosamide, ivermectin, albendazole derivatives, nitazoxanide and pyrimethamine, have been deeply investigated and widely applied in treating various parasitic diseases for a long time. Generally, their pharmacokinetic and pharmacodynamic properties are well understood, while the side effects are roughly acceptable. Scientists noticed that some of these agents have anticancer potentials and explored the underlying mechanisms to achieve drug repurposing. Recent studies show that these agents inhibit cancer progression via multiple interesting ways, inducing ferroptosis induction, autophagy regulation, mitochondrial disturbance, immunoregulation, and metabolic disruption. In this review, we summarize the recent advancement in uncovering antiparasitic drugs' anticancer properties from the perspective of their pharmacological targets. Instead of paying attention to the previously discovered mechanisms, we focus more on newly emerging ones that are worth noticing. While most investigations are focusing on the mechanisms of their antiparasitic effect, more in vivo exploration in clinical trials in the future is necessary. Moreover, we also paid attention to what limits the clinical application of these agents. For some of these agents like ARTs and niclosamide, drug modification, novel delivery system invention, or drug combination are strongly recommended for future exploration.
Collapse
Affiliation(s)
- Haoyang Huang
- Department of Clinical Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Qing He
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China.,CAEA Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, 100048, People's Republic of China
| | - Binghua Guo
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Xudong Xu
- Department of Clinical Medicine, School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Yinjuan Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China.,CAEA Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, 100048, People's Republic of China
| | - Xuerong Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China.,CAEA Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, 100048, People's Republic of China
| |
Collapse
|
44
|
Meng Y, Ma N, Lyu H, Wong YK, Zhang X, Zhu Y, Gao P, Sun P, Song Y, Lin L, Wang J. Recent pharmacological advances in the repurposing of artemisinin drugs. Med Res Rev 2021; 41:3156-3181. [PMID: 34148245 DOI: 10.1002/med.21837] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/27/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022]
Abstract
Artemisinins are a family of sesquiterpene lactones originally derived from the sweet wormwood (Artemisia annua). Beyond their well-characterized role as frontline antimalarial drugs, artemisinins have also received increased attention for other potential pharmaceutical effects, which include antiviral, antiparsitic, antifungal, anti-inflammatory, and anticancer activities. With concerted efforts in further preclinical and clinical studies, artemisinin-based drugs have the potential to be viable treatments for a great variety of human diseases. Here, we provide a comprehensive update on recent reports of pharmacological actions and applications of artemisinins outside of their better-known antimalarial role and highlight their potential therapeutic viability for various diseases.
Collapse
Affiliation(s)
- Yuqing Meng
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Ma
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haining Lyu
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yin Kwan Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xing Zhang
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongping Zhu
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Gao
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Sun
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yali Song
- Center for Reproductive Medicine, Dongguan Maternal And Child Health Care Hospital, Southern Medical University, Dongguan, China
| | - Lizhu Lin
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jigang Wang
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
45
|
Feng J, Mansouripour A, Xi Z, Zhang L, Xu G, Zhou H, Xu H. Nujiangexanthone A Inhibits Cervical Cancer Cell Proliferation by Promoting Mitophagy. Molecules 2021; 26:2858. [PMID: 34065886 PMCID: PMC8150697 DOI: 10.3390/molecules26102858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/29/2021] [Accepted: 05/06/2021] [Indexed: 12/22/2022] Open
Abstract
Nujiangexanthone A (NJXA), a bioactive component isolated from the leaves of Garcinia nujiangensis, has been reported to exhibit anti-inflammatory, antioxidant, and antitumor effects. Our previous work has shown that NJXA induced G0/1 arrest and apoptosis, thus suppressing cervical cancer cell growth. The present study provides new evidence that NJXA can induce cell death in HeLa cells by promoting mitophagy. We first identified that NJXA triggered GFP-LC3 and YFP-Parkin puncta accumulation, which are biomarkers of mitophagy. Moreover, NJXA degraded the mitochondrial membrane proteins Tom20 and Tim23 and mitochondrial fusion proteins MFN1 and MFN2, downregulated Parkin, and stabilized PINK1. Additionally, we revealed that NJXA induced lysosome degradation and colocalization of mitochondria and autophagosomes, which was attenuated by knocking down ATG7, the key regulator of mitophagy. Furthermore, since mitophagy is induced under starvation conditions, we detected the cytotoxic effect of NJXA in nutrient-deprived HeLa cells and observed better cytotoxicity. Taken together, our work contributes to the further clarification of the mechanism by which NJXA inhibits cervical cancer cell proliferation and provides evidence that NJXA has the potential to develop anticancer drugs.
Collapse
Affiliation(s)
- Jiling Feng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China;
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China; (A.M.); (Z.X.); (L.Z.)
| | - Anahitasadat Mansouripour
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China; (A.M.); (Z.X.); (L.Z.)
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China; (A.M.); (Z.X.); (L.Z.)
| | - Li Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China; (A.M.); (Z.X.); (L.Z.)
| | - Gang Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China;
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China;
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China; (A.M.); (Z.X.); (L.Z.)
| | - Hongxi Xu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, No. 528, Zhangheng Road, Shanghai 201203, China;
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Shanghai 201203, China; (A.M.); (Z.X.); (L.Z.)
| |
Collapse
|
46
|
Raj SD, Fann DY, Wong E, Kennedy BK. Natural products as geroprotectors: An autophagy perspective. Med Res Rev 2021; 41:3118-3155. [PMID: 33973253 DOI: 10.1002/med.21815] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/09/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022]
Abstract
Over the past decade, significant attention has been given to repurposing Food and Drug Administration approved drugs to treat age-related diseases. In contrast, less consideration has been given to natural bioactive compounds. Consequently, there have been limited attempts to translate these compounds. Autophagy is a fundamental biological pathway linked to aging, and numerous strategies to enhance autophagy have been shown to extend lifespan. Interestingly, there are a number of natural products that are reported to modulate autophagy, and here we describe a number of them that activate autophagy through diverse molecular and cellular mechanisms. Among these, Urolithin A, Spermidine, Resveratrol, Fatty Acids and Phospholipids, Trehalose and Lithium are featured in detail. Finally, we outline possible strategies to optimise and increase the translatability of natural products, with the overall aim of delaying the ageing process and improving human healthspan.
Collapse
Affiliation(s)
- Stephen D Raj
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre For Healthy Longevity, National University Health System, National University of Singapore, Singapore
| | - David Y Fann
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre For Healthy Longevity, National University Health System, National University of Singapore, Singapore
| | - Esther Wong
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre For Healthy Longevity, National University Health System, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Brian K Kennedy
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre For Healthy Longevity, National University Health System, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Agency for Science, Technology and Research (A*STAR), Singapore Institute for Clinical Sciences, Singapore
| |
Collapse
|
47
|
Shu YH, Yuan HH, Xu MT, Hong YT, Gao CC, Wu ZP, Han HT, Sun X, Gao RL, Yang SF, Li SX, Tian JK, Zhang JB. A novel Diels-Alder adduct of mulberry leaves exerts anticancer effect through autophagy-mediated cell death. Acta Pharmacol Sin 2021; 42:780-790. [PMID: 32814819 PMCID: PMC8115316 DOI: 10.1038/s41401-020-0492-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/22/2020] [Indexed: 02/01/2023] Open
Abstract
Guangsangon E (GSE) is a novel Diels-Alder adduct isolated from leaves of Morus alba L, a traditional Chinese medicine widely applied in respiratory diseases. It is reported that GSE has cytotoxic effect on cancer cells. In our research, we investigated its anticancer effect on respiratory cancer and revealed that GSE induces autophagy and apoptosis in lung and nasopharyngeal cancer cells. We first observed that GSE inhibits cell proliferation and induces apoptosis in A549 and CNE1 cells. Meanwhile, the upregulation of autophagosome marker LC3 and increased formation of GFP-LC3 puncta demonstrates the induction of autophagy in GSE-treated cells. Moreover, GSE increases the autophagy flux by enhancing lysosomal activity and the fusion of autophagosomes and lysosomes. Next, we investigated that endoplasmic reticulum (ER) stress is involved in autophagy induction by GSE. GSE activates the ER stress through reactive oxygen species (ROS) accumulation, which can be blocked by ROS scavenger NAC. Finally, inhibition of autophagy attenuates GSE-caused cell death, termed as "autophagy-mediated cell death." Taken together, we revealed the molecular mechanism of GSE against respiratory cancer, which demonstrates great potential of GSE in the treatment of representative cancer.
Collapse
Affiliation(s)
- Yu-Han Shu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310058, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Hua-Hua Yuan
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310058, China
| | - Meng-Ting Xu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310058, China
| | - Ye-Ting Hong
- Hangzhou Medical College, Hangzhou, 310053, China
| | - Cheng-Cheng Gao
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310058, China
| | - Zhi-Pan Wu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310058, China
| | - Hao-Te Han
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310058, China
| | - Xin Sun
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Rui-Lan Gao
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Si-Fu Yang
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Shou-Xin Li
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang-Malaysia Joint Research Center for Traditional Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jing-Kui Tian
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang-Malaysia Joint Research Center for Traditional Medicine, Zhejiang University, Hangzhou, 310058, China.
| | - Jian-Bin Zhang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
48
|
Artesunate-induced ATG5-related autophagy enhances the cytotoxicity of NK92 cells on endometrial cancer cells via interactions between CD155 and CD226/TIGIT. Int Immunopharmacol 2021; 97:107705. [PMID: 33933849 DOI: 10.1016/j.intimp.2021.107705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
Uterine corpus endometrial carcinoma (UCEC) is the most prevalent gynecologic cancer in developed countries and lacks efficient therapeutic strategies. Artesunate (ART), a well-modified derivate of artemisinin, exerts potent anti-cancer effects apart from its classical anti-malaria feature. Autophagy is a universal double-edged process in cell survival, and CD155 is a novel immune checkpoint highly expressed in numerous cancers. However, the relationships among ART, autophagy, and CD155 remain unclear in UCEC. In this study, we discovered that ART not only inhibited proliferation and migration, promoted apoptosis, but also induced autophagy in UCEC cells. Meanwhile, ART-induced autophagy elevated the level of CD155 in UCEC cells, thereby enhancing the cytotoxicity of natural killer cell line (NK92) by modulating the interactions between CD155 and its receptors in NK92 cells via upregulation of co-stimulator CD226 and downregulation of co-inhibitor TIGIT. Additionally, ART regulated CD155 partially via ATG5, and knockdown of ATG5 dampened the expression of CD155 in UCEC cells, thus decreasing the cytotoxicity of NK92 cells. Therefore, this study demonstrated the dual anti-cancer effects of ART as it could induce cell-killing directly and indirectly, which provides novel insights into the anti-cancer mechanisms of ART on UCEC.
Collapse
|
49
|
Pereira GJDS, Leão AHFF, Erustes AG, Morais IBDM, Vrechi TADM, Zamarioli LDS, Pereira CAS, Marchioro LDO, Sperandio LP, Lins ÍVF, Piacentini M, Fimia GM, Reckziegel P, Smaili SS, Bincoletto C. Pharmacological Modulators of Autophagy as a Potential Strategy for the Treatment of COVID-19. Int J Mol Sci 2021; 22:4067. [PMID: 33920748 PMCID: PMC8071111 DOI: 10.3390/ijms22084067] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
The family of coronaviruses (CoVs) uses the autophagy machinery of host cells to promote their growth and replication; thus, this process stands out as a potential target to combat COVID-19. Considering the different roles of autophagy during viral infection, including SARS-CoV-2 infection, in this review, we discuss several clinically used drugs that have effects at different stages of autophagy. Among them, we mention (1) lysosomotropic agents, which can prevent CoVs infection by alkalinizing the acid pH in the endolysosomal system, such as chloroquine and hydroxychloroquine, azithromycin, artemisinins, two-pore channel modulators and imatinib; (2) protease inhibitors that can inhibit the proteolytic cleavage of the spike CoVs protein, which is necessary for viral entry into host cells, such as camostat mesylate, lopinavir, umifenovir and teicoplanin and (3) modulators of PI3K/AKT/mTOR signaling pathways, such as rapamycin, heparin, glucocorticoids, angiotensin-converting enzyme inhibitors (IECAs) and cannabidiol. Thus, this review aims to highlight and discuss autophagy-related drugs for COVID-19, from in vitro to in vivo studies. We identified specific compounds that may modulate autophagy and exhibit antiviral properties. We hope that research initiatives and efforts will identify novel or "off-label" drugs that can be used to effectively treat patients infected with SARS-CoV-2, reducing the risk of mortality.
Collapse
Affiliation(s)
- Gustavo José da Silva Pereira
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Anderson Henrique França Figueredo Leão
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Adolfo Garcia Erustes
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Ingrid Beatriz de Melo Morais
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Talita Aparecida de Moraes Vrechi
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Lucas dos Santos Zamarioli
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Cássia Arruda Souza Pereira
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Laís de Oliveira Marchioro
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Letícia Paulino Sperandio
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Ísis Valeska Freire Lins
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Mauro Piacentini
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy;
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS ‘La Zaro Spallanzani’, 00149 Rome, Italy;
| | - Gian Maria Fimia
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS ‘La Zaro Spallanzani’, 00149 Rome, Italy;
- Department of Molecular Medicine, University of Rome La Sapienza, 00185 Rome, Italy
| | - Patrícia Reckziegel
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Soraya Soubhi Smaili
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| | - Claudia Bincoletto
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), 04044-020 São Paulo, Brazil; (A.H.F.F.L.); (A.G.E.); (I.B.d.M.M.); (T.A.d.M.V.); (L.d.S.Z.); (C.A.S.P.); (L.d.O.M.); (L.P.S.); (Í.V.F.L.); (P.R.); (S.S.S.); (C.B.)
| |
Collapse
|
50
|
Khanal P. Antimalarial and anticancer properties of artesunate and other artemisinins: current development. MONATSHEFTE FUR CHEMIE 2021; 152:387-400. [PMID: 33814617 PMCID: PMC8008344 DOI: 10.1007/s00706-021-02759-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
This review provides a recent perspective of artesunate and other artemisinins as antimalarial drugs and their uses in cancer therapy. Artesunate is an artemisinin derivative. Artemisinin is extracted from the plant Artemisia annua. Artemisinin and its derivatives have been the most useful drug for malarial treatment in human history. The artesunate has an advantage of a hydrophilic group over other artemisinins which makes it a more potent drug. On the industrial scale, artemisinins are synthesized in semisynthetic ways. The 1,2,4-endoperoxide bridge of artemisinins is responsible for the drug's antimalarial activity. There is the emergence of artemisinin resistance on Plasmodium falciparum and pieces of evidence suggest that it is mainly due to the mutation at Kelch13 protein of P. falciparum. Clinical trial data show that the artesunate is more favorable than quinine and other artemisinins to treat patients with severe malaria. Pieces of evidence indicate that artemisinins can be developed as anticancer drugs. The mechanism of actions on how artemisinins act as an anticancer drug involves oxidative stress, DNA damage and repair, and various types of cell deaths. GRAPHIC ABSTRACT
Collapse
Affiliation(s)
- Pitambar Khanal
- Nagarik College, Tribhuvan University, Gaidakot-2, Nawalparasi Purva, Gandaki, Nepal
| |
Collapse
|