1
|
Hu H, Zhang H, Zhong R, Yang Y, Huang C, Chen J, Liang L, Chen Y, Liu Y. Synthesis, RNA-sequence and evaluation of anticancer efficacy of ruthenium(II) polypyridyl complexes toward HepG2 cells. J Inorg Biochem 2023; 244:112230. [PMID: 37084581 DOI: 10.1016/j.jinorgbio.2023.112230] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/05/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
In this article, four new Ru(II) complexes [Ru(dmbpy)2(TFBIP)](PF6)2 (dmbpy = 4,4'-dimethyl-2,2'-bipyridine, TFPIP = 2-(4'-trifluoromethyl)-[1,1'-biphenyl]-4-yl)-1H-imidazo[4,5-f][1,10]phenanthroline) (Ru1), [Ru(bpy)2(TFBIP)](PF6)2 (bpy = 2,2'-bipyridine) (Ru2), [Ru(phen)2(TFBIP)](PF6)2 (phen = 1,10-phenanthroline) (Ru3) and [Ru(dmp)2(TFBIP)](PF6)2 (dmp = 2,9-dimethyl-1,10-phenanthroline) (Ru4) were synthesized and characterized by elemental analysis, HRMS, IR, 1H NMR, 13C NMR and 19F NMR. The in vitro anticancer effect of the complexes on HepG2, A549, B16, HeLa, BEL-7402 and non-cancer LO2 cells was screened using 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method. The results illustrate that the complexes display moderate anticancer activity. Apoptotic assay with Annexin V/PI double staining method indicated that complexes induce apoptosis in HepG2 cells. Also, the complexes interfere with the mitochondrial functions, accompanied by the production of intracellular ROS as well as a reduction of mitochondrial membrane potential. The results obtained from the western blot demonstrated that the complexes upregulate pro-apoptotic Bax and downregulate anti-apoptotic Bcl-2, which further activates caspase 3 and promotes the cleavage of PARP. RNA-sequence showed that the complexes upregulate the expression of 40 genes and downregulate 66 genes. Antitumour in vivo demonstrated that Ru1 inhibits the tumor growth with a high inhibitory rate of 51.19%. Taken together, these results revealed that complexes Ru1, Ru2, Ru3 and Ru4 induce cell death in HepG2 cells via autophagy and a ROS-mediated mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Huiyan Hu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huiwen Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Ruitong Zhong
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yan Yang
- Department of Pharmacy, Guangdong Second Provincial General Hospital, 510317, PR China.
| | - Chunxia Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jing Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yichuan Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
2
|
Dong CL, Zhu F, Du YZ, Lu MX. Depending on different apoptosis pathways, the effector Cscaspase-3 in Chilo suppressalis exposed to temperature and parasitic stress was induced. Int J Biol Macromol 2023; 238:124270. [PMID: 37003373 DOI: 10.1016/j.ijbiomac.2023.124270] [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: 02/21/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Apoptosis is a form of programmed cell death (PCD) that is largely triggered by caspases through both the mitochondria-dependent and mitochondria-independent pathways. The rice stem borer, Chilo suppressalis, serves as an economically important pest of rice, which is often suffered by temperature and parasitic stress under natural conditions. In the present study, effector Cscaspase-3 encoding caspase was obtained from the rice pest Chilo suppressalis. CsCaspase-3 possesses p20 and p10 subunits, two active sites, four substrate-binding sites, and two cleavage motifs. Real-time quantitative PCR showed that Cscaspase-3 was expressed at maximal levels in hemocytes; furthermore, transcription was most highly in female adults. Expression of Cscaspase-3 was induced by hot and cold temperatures, with the highest expression at 39 °C. Cscaspase-3 expression was also significantly induced at 10 h, 2 d, 5 d, and 7 d of parasitism. Flow cytometry results showed that both temperature and parasitism trigger apoptosis, but only parasitism induces apoptosis via the mitochondrial apoptosis pathway in C. suppressalis. RNAi-mediated silencing of Cscaspase-3 expression reduced C. suppressalis survival at -3 °C. This study provides a foundation for further studies of caspases in insects during biotic and abiotic stress.
Collapse
Affiliation(s)
- Chuan-Lei Dong
- College of Plant Protection & Institute of Applied Entomology, Yangzhou University, Yangzhou 225009, China
| | - Feng Zhu
- Plant Protection and Quarantine Station of Jiangsu Province, Nanjing 210000, PR China
| | - Yu-Zhou Du
- College of Plant Protection & Institute of Applied Entomology, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education, Yangzhou University, Yangzhou, China.
| | - Ming-Xing Lu
- College of Plant Protection & Institute of Applied Entomology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
3
|
Zare A, Afshar A, Khoradmehr A, Baghban N, Mohebbi G, Barmak A, Daneshi A, Bargahi A, Nabipour I, Almasi-Turk S, Arandian A, Zibaii MI, Latifi H, Tamadon A. Chemical Compositions and Experimental and Computational Modeling of the Anticancer Effects of Cnidocyte Venoms of Jellyfish Cassiopea andromeda and Catostylus mosaicus on Human Adenocarcinoma A549 Cells. Mar Drugs 2023; 21:md21030168. [PMID: 36976217 PMCID: PMC10057638 DOI: 10.3390/md21030168] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 03/09/2023] Open
Abstract
Nowadays, major attention is being paid to curing different types of cancers and is focused on natural resources, including oceans and marine environments. Jellyfish are marine animals with the ability to utilize their venom in order to both feed and defend. Prior studies have displayed the anticancer capabilities of various jellyfish. Hence, we examined the anticancer features of the venom of Cassiopea andromeda and Catostylus mosaicus in an in vitro situation against the human pulmonary adenocarcinoma (A549) cancer cell line. The MTT assay demonstrated that both mentioned venoms have anti-tumoral ability in a dose-dependent manner. Western blot analysis proved that both venoms can increase some pro-apoptotic factors and reduce some anti-apoptotic molecules that lead to the inducing of apoptosis in A549 cells. GC/MS analysis demonstrated some compounds with biological effects, including anti-inflammatory, antioxidant and anti-cancer activities. Molecular docking and molecular dynamic showed the best position of each biologically active component on the different death receptors, which are involved in the process of apoptosis in A549 cells. Ultimately, this study has proven that both venoms of C. andromeda and C. mosaicus have the capability to suppress A549 cells in an in vitro condition and they might be utilized in order to design and develop brand new anticancer agents in the near future.
Collapse
Affiliation(s)
- Afshin Zare
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr 75, Iran
| | - Alireza Afshar
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr 75, Iran
- PerciaVista R&D Co., Shiraz 73, Iran
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 73, Iran
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 73, Iran
| | - Neda Baghban
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 73, Iran
| | - Gholamhossein Mohebbi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 73, Iran
| | - Alireza Barmak
- Food Lab, Bushehr University of Medical Sciences, Bushehr 73, Iran
| | - Adel Daneshi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 73, Iran
| | - Afshar Bargahi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 73, Iran
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 73, Iran
| | - Sahar Almasi-Turk
- Department of Anatomical Sciences, School of Medicine, Bushehr University of Medical Sciences, Bushehr 73, Iran
- Correspondence: (S.A.-T.); (A.T.); Tel.: +98-77-3332-0657 (S.A.-T.); +98-21-2842-6122 (A.T.)
| | - Alireza Arandian
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran 11, Iran
| | | | - Hamid Latifi
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran 11, Iran
- Department of Physics, Shahid Beheshti University, Tehran 11, Iran
| | - Amin Tamadon
- PerciaVista R&D Co., Shiraz 73, Iran
- Correspondence: (S.A.-T.); (A.T.); Tel.: +98-77-3332-0657 (S.A.-T.); +98-21-2842-6122 (A.T.)
| |
Collapse
|
4
|
Nardoguaianone L Isolated from Nardostachys jatamansi Improved the Effect of Gemcitabine Chemotherapy via Regulating AGE Signaling Pathway in SW1990 Cells. Molecules 2022; 27:molecules27206849. [PMID: 36296442 PMCID: PMC9610730 DOI: 10.3390/molecules27206849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related death worldwide and is known as “the king of cancers”. Currently, gemcitabine (GEM) as the clinical drug of choice for chemotherapy of advanced pancreatic cancer has poor drug sensitivity and ineffective chemotherapy. Nardoguaianone L (G-6) is a novel guaiane-type sesquiterpenoid isolated from Nardostachys jatamansi DC., and it exhibits anti-tumor activity. Based on the newly discovered G-6 with anti-pancreatic cancer activity in our laboratory, this paper aimed to evaluate the potential value of the combination of G-6 and GEM in SW1990 cells, including cell viability, cell apoptosis, colony assay and tandem mass tags (TMT) marker-based proteomic technology. These results showed that G-6 combined with GEM significantly inhibited cell viability, and the effect was more obvious than that with single drug. In addition, the use of TMT marker-based proteomic technology demonstrated that the AGE-RAGE signaling pathway was activated after medication-combination. Furthermore, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) assays were used to validate the proteomic results. Finally, apoptosis was detected by flow cytometry. In conclusion, G-6 combined with GEM induced an increase in ROS level and a decrease in MMP in SW1990 cells through the AGE-RAGE signaling pathway, ultimately leading to apoptosis. G-6 improved the effect of GEM chemotherapy and may be used as a potential combination therapy for pancreatic cancer.
Collapse
|
5
|
Zhao P, Qiu H, Wei Q, Li Y, Gao L, Zhao P. Anti-tumor effect of novel amino acid Schiff base nickel (II) complexes on oral squamous cell carcinoma cells (CAL-27) in vitro. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00255-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
6
|
Cyclometalated Ru(II) β-carboline complexes induce cell cycle arrest and apoptosis in human HeLa cervical cancer cells via suppressing ERK and Akt signaling. J Biol Inorg Chem 2021; 26:793-808. [PMID: 34459988 DOI: 10.1007/s00775-021-01894-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
Two new cyclometalated Ru(II)-β-carboline complexes, [Ru(dmb)2(Cl-Ph-βC)](PF6) (dmb = 4,4'-dimethyl-2,2'-bipyridine; Cl-Ph-βC = Cl-phenyl-9H-pyrido[3,4-b]indole; RuβC-3) and [Ru(bpy)2(Cl-Ph-βC)](PF6) (bpy = 2,2'-bipyridine; RuβC-4) were synthesized and characterized. The Ru(II) complexes display high cytotoxicity against HeLa cells, the stabilized human cervical cancer cell, with IC50 values of 3.2 ± 0.4 μM (RuβC-3) and 4.1 ± 0.6 μM (RuβC-4), which were considerably lower than that of non-cyclometalated Ru(II)-β-carboline complex [Ru(bpy)2(1-Py-βC)] (PF6)2 (61.2 ± 3.9 μM) by 19- and 15-folds, respectively. The mechanism studies indicated that both Ru(II) complexes could significantly inhibit HeLa cell migration and invasion, and effectively induce G0/G1 cell cycle arrest. The new Ru(II) complexes could also trigger apoptosis through activating caspase-3 and poly (ADP-ribose) polymerase (PARP), increasing the Bax/Bcl-2 ratio, enhancing reactive oxygen species (ROS) generation, decreasing mitochondrial membrane potential (MMP), and inducing cytochrome c release from mitochondria. Further research revealed that RuβC-3 could deactivate the ERK/Akt signaling pathway thus inhibiting HeLa cell invasion and migration, and inducing apoptosis. In addition, RuβC-3-induced apoptosis in HeLa cells was closely associated with the increase of intracellular ROS levels, which may act as upstream factors to regulate ERK and Akt pathways. More importantly, RuβC-3 exhibited low toxicity on both normal BEAS-2B cells in vitro and zebrafish embryos in vivo. Consequently, the developed Ru(II) complexes have great potential on developing novel low-toxic anticancer drugs.
Collapse
|
7
|
Li Q, Ma Q, Cheng J, Zhou X, Pu W, Zhong X, Guo X. Dihydroartemisinin as a Sensitizing Agent in Cancer Therapies. Onco Targets Ther 2021; 14:2563-2573. [PMID: 33880035 PMCID: PMC8053502 DOI: 10.2147/ott.s297785] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/18/2021] [Indexed: 01/03/2023] Open
Abstract
Cancer is one of the major threats to human health. Although humans have struggled with cancer for decades, the efficacy of treatments for most tumors is still very limited. Dihydroartemisinin (DHA) is a derivative of artemisinin, a first-line antimalarial drug originally developed in China. Beyond the anti-malarial effect, DHA has also been reported to show anti-inflammatory, anti-parasitosis, and immune-modulating properties in vitro and in vivo. Furthermore, an increasing number of studies report that DHA possesses anticancer activities on a wide range of cancer types both in vitro and in vivo, as well as enhances the efficacy of chemotherapy, targeted therapy, and even radiotherapy. However, the mechanisms of DHA on different tumors differ in various ways. In this review, we intend to summarize how DHA sensitizes cancer cells to anti-cancer therapies, highlight its molecular mechanisms and pharmacological effects in vitro and in vivo as well as in current clinical trials, and discuss potential issues concerning DHA. Hopefully, more attention will be paid to DHA as a sensitizer for cancer therapy in the future.
Collapse
Affiliation(s)
- Qingrong Li
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Qiang Ma
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Jibing Cheng
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xi Zhou
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Wenjie Pu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xiaowu Zhong
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| |
Collapse
|
8
|
Zamani F, Samiei F, Mousavi Z, Azari MR, Seydi E, Pourahmad J. Apigenin ameliorates oxidative stress and mitochondrial damage induced by multiwall carbon nanotubes in rat kidney mitochondria. J Biochem Mol Toxicol 2021; 35:1-7. [PMID: 33724625 DOI: 10.1002/jbt.22762] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/01/2020] [Accepted: 03/02/2021] [Indexed: 12/19/2022]
Abstract
The toxicity of carbon nanotubes (CNTs) toward the mitochondria of the kidney is not fully recognized and still needs further research. Apigenin (APG) is known as a flavonoid compound and natural antioxidant. The purpose of this study was to assess the ameliorative role of APG against multiwall CNT (MWCNT)-induced kidney toxicity in rats. The animals were administrated with APG (10 mg/kg) for 2 weeks and then were exposed to MWCNTs (5 mg/m3 ) in pure and impure forms (10 and 100 nm) for 5 h/day and 5 days/week. Then, mitochondria were isolated from the kidney tissue and mitochondrial toxicity parameters were measured. Decreases in succinate dehydrogenase activity have been reported in all groups exposed to MWCNTs. Results indicated that MWCNTs in both forms and sizes were able to increase the generation of reactive oxygen species, decline mitochondrial membrane potential, induce mitochondrial swelling, and release cytochrome c in isolated kidney mitochondria. The pretreatment of APG decreased all the abovementioned mitochondrial damage and oxidative stress parameters induced by both pure and impure MWCNTs. Our results showed that MWCNTs have the ability to enter the body, subsequently, cross cellular barriers, and reach the kidney as a sensitive organ, which can result in mitochondrial damage in kidney cells including renal tubular cells. In addition, APG can be an effective nutritional antioxidant regimen against MWCNT-induced kidney damage.
Collapse
Affiliation(s)
- Fatemeh Zamani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Samiei
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Mousavi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mansour Rezazadeh Azari
- School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Enayatollah Seydi
- Department of Occupational Health and Safety Engineering, School of Health, Alborz University of Medical Sciences, Karaj, Iran.,Research Center for Health, Safety, and Environment, Alborz University of Medical Sciences, Karaj, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Qin LQ, Liang CJ, Zhou Z, Qin QP, Wei ZZ, Tan MX, Liang H. Mitochondria-localizing curcumin-cryptolepine Zn(II) complexes and their antitumor activity. Bioorg Med Chem 2021; 30:115948. [PMID: 33360578 DOI: 10.1016/j.bmc.2020.115948] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 01/08/2023]
Abstract
Many metal complexes are potent candidates as mitochondrial-targeting agents. In this study, four novel Zn(II) complexes, [Zn(BPQA)Cl2] (Zn1), [Zn(BPQA)(Curc)]Cl (Zn2), [Zn(PQA)Cl2] (Zn3), and [Zn(PQA)(Curc)]Cl (Zn4), containing N,N-bis(pyridin-2-ylmethyl)benzofuro[3,2-b]quinolin-11-amine (BPQA), N-(pyridin-2-ylmethyl)benzofuro[3,2-b]quinolin-11-amine (PQA), and curcumin (H-Curc) were synthesized. An MTT assay showed that Zn1-Zn4 had strong anticancer activities against SK-OV-3/DDP and T-24 tumor cells with IC50 values of 0.03-6.19 μM. Importantly, Zn1 and Zn2 displayed low toxicities against normal HL-7702 cells. Mechanism experiments demonstrated that probe Zn2 showed appreciable fluorescence in the red region of the spectrum, and substantial accumulation of Zn2 occurred in the mitochondria after treatment, indicating increases in Ca2+ and reactive oxygen species levels, loss of the mitochondrial membrane potential, and consequent induction of mitochondrial dysfunction at low concentrations. In addition, the probe Zn2 effectively (50.7%) inhibited the growth of T-24 bladder tumor cells in vivo. The probe Zn2 shows potential for use in cancer therapy while retaining the H-Curc as an imaging probe.
Collapse
Affiliation(s)
- Li-Qin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Chun-Jie Liang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Zhen Zhou
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China.
| | - Zu-Zhuang Wei
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China.
| | - Ming-Xiong Tan
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China.
| |
Collapse
|
10
|
Xu C, Zhang H, Mu L, Yang X. Artemisinins as Anticancer Drugs: Novel Therapeutic Approaches, Molecular Mechanisms, and Clinical Trials. Front Pharmacol 2020; 11:529881. [PMID: 33117153 PMCID: PMC7573816 DOI: 10.3389/fphar.2020.529881] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 09/16/2020] [Indexed: 12/26/2022] Open
Abstract
Artemisinin and its derivatives have shown broad-spectrum antitumor activities in vitro and in vivo. Furthermore, outcomes from a limited number of clinical trials provide encouraging evidence for their excellent antitumor activities. However, some problems such as poor solubility, toxicity and controversial mechanisms of action hamper their use as effective antitumor agents in the clinic. In order to accelerate the use of ARTs in the clinic, researchers have recently developed novel therapeutic approaches including developing novel derivatives, manufacturing novel nano-formulations, and combining ARTs with other drugs for cancer therapy. The related mechanisms of action were explored. This review describes ARTs used to induce non-apoptotic cell death containing oncosis, autophagy, and ferroptosis. Moreover, it highlights the ARTs-caused effects on cancer metabolism, immunosuppression and cancer stem cells and discusses clinical trials of ARTs used to treat cancer. The review provides additional insight into the molecular mechanism of action of ARTs and their considerable clinical potential.
Collapse
Affiliation(s)
- Cangcang Xu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Huihui Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Lingli Mu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| |
Collapse
|
11
|
Li Y, Dong J, Zhao P, Hu P, Yang D, Gao L, Li L. Synthesis of Amino Acid Schiff Base Nickel (II) Complexes as Potential Anticancer Drugs In Vitro. Bioinorg Chem Appl 2020; 2020:8834859. [PMID: 33061947 PMCID: PMC7542481 DOI: 10.1155/2020/8834859] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/18/2020] [Indexed: 11/22/2022] Open
Abstract
Three hexacoordinated octahedral nickel (II) complexes, [Ni (Trp-sal) (phen) (CH3OH)] (1), [Ni (Trp-o-van) (phen) (CH3OH)]•2CH3OH (2), and [Ni (Trp-naph) (phen) (CH3OH)] (3) (where Trp-sal = Schiff base derived from tryptophan and salicylaldehyde, Trp-o-van = Schiff base derived from tryptophan and o-vanillin, Trp-naph = Schiff base derived from tryptophan and 2-hydroxy-1-naphthaldehyde, phen = 1, 10-phenanthroline), have been synthesized and characterized as potential anticancer agents. Details of structural study of these complexes using single-crystal X-ray crystallography showed that distorted octahedral environment around nickel (II) ion has been satisfied by three nitrogen atoms and three oxygen atoms. All these complexes displayed moderate cytotoxicity toward esophageal cancer cell line Eca-109 with the IC50 values of 23.95 ± 2.54 μM for 1, 18.14 ± 2.39 μM for 2, and 21.89 ± 3.19 μM for 3. Antitumor mechanism studies showed that complex 2 can increase the autophagy, reactive oxygen species (ROS) levels, and decrease the mitochondrial membrane potential remarkably in a dose-dependent manner in the Eca-109 cells. Complex 2 can cause cell cycle arrest in the G2/M phase. Additionally, complex 2 can regulate the Bcl-2 family and autophagy-related proteins.
Collapse
Affiliation(s)
- Yang Li
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Jianfang Dong
- Department of Material Science, Shandong Polytechnic Technician College, Liaocheng 252000, China
| | - Peiran Zhao
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Ping Hu
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Dawei Yang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Lei Gao
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Lianzhi Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
12
|
Cheong DHJ, Tan DWS, Wong FWS, Tran T. Anti-malarial drug, artemisinin and its derivatives for the treatment of respiratory diseases. Pharmacol Res 2020; 158:104901. [PMID: 32405226 PMCID: PMC7217791 DOI: 10.1016/j.phrs.2020.104901] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023]
Abstract
Artemisinins are sesquiterpene lactones with a peroxide moiety that are isolated from the herb Artemisia annua. It has been used for centuries for the treatment of fever and chills, and has been recently approved for the treatment of malaria due to its endoperoxidase properties. Progressively, research has found that artemisinins displayed multiple pharmacological actions against inflammation, viral infections, and cell and tumour proliferation, making it effective against diseases. Moreover, it has displayed a relatively safe toxicity profile. The use of artemisinins against different respiratory diseases has been investigated in lung cancer models and inflammatory-driven respiratory disorders. These studies revealed the ability of artemisinins in attenuating proliferation, inflammation, invasion, and metastasis, and in inducing apoptosis. Artemisinins can regulate the expression of pro-inflammatory cytokines, nuclear factor-kappa B (NF-κB), matrix metalloproteinases (MMPs), vascular endothelial growth factor (VEGF), promote cell cycle arrest, drive reactive oxygen species (ROS) production and induce Bak or Bax-dependent or independent apoptosis. In this review, we aim to provide a comprehensive update of the current knowledge of the effects of artemisinins in relation to respiratory diseases to identify gaps that need to be filled in the course of repurposing artemisinins for the treatment of respiratory diseases. In addition, we postulate whether artemisinins can also be repurposed for the treatment of COVID-19 given its anti-viral and anti-inflammatory properties.
Collapse
Affiliation(s)
- Dorothy H J Cheong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore
| | - Daniel W S Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Fred W S Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; Immunology Program, Life Science Institute, National University of Singapore, 117456, Singapore; Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, 138602, Singapore
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore.
| |
Collapse
|
13
|
Gao J, Ma F, Wang X, Li G. Combination of dihydroartemisinin and resveratrol effectively inhibits cancer cell migrationviaregulation of the DLC1/TCTP/Cdc42 pathway. Food Funct 2020; 11:9573-9584. [DOI: 10.1039/d0fo00996b] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mechanism of DHA combined with RES in inhibition of cancer cell migration by DLC1/TCTP/Cdc42 signaling.
Collapse
Affiliation(s)
- Junying Gao
- Shandong Provincial Key Laboratory of Animal Resistant Biology
- School of Life Sciences
- Shandong Normal University
- Jinan
- China
| | - Fengqiu Ma
- Shandong Provincial Key Laboratory of Animal Resistant Biology
- School of Life Sciences
- Shandong Normal University
- Jinan
- China
| | - Xingjie Wang
- Shandong Provincial Key Laboratory of Animal Resistant Biology
- School of Life Sciences
- Shandong Normal University
- Jinan
- China
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant Biology
- School of Life Sciences
- Shandong Normal University
- Jinan
- China
| |
Collapse
|
14
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 280] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
15
|
Ahmad A, Kumari P, Ahmad M. Apigenin attenuates edifenphos-induced toxicity by modulating ROS-mediated oxidative stress, mitochondrial dysfunction and caspase signal pathway in rat liver and kidney. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2019; 159:163-172. [PMID: 31400778 DOI: 10.1016/j.pestbp.2019.06.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/08/2019] [Accepted: 06/21/2019] [Indexed: 06/10/2023]
Abstract
Edifenphos (EDF) (O-ethyl-S, S-diphenyldithiophosphate) is an organophosphate pesticide that is extensively used as a fungicide in agricultural rice fields. However, EDF accumulated in various agricultural products and caused potential health hazards to human and other living organisms. Therefore, the present study was investigated to evaluate the ameliorative role of apigenin (APG); a natural antioxidant against EDF-induced hepato-renal toxicity in rats. Six groups with five male Wistar rats each, were used for this purpose; these groups included the control group (A) that received corn oil; (B) 10 mg/kg APG; (C) 10 mg/kg EDF; (D) 25 mg/kg EDF; (E) 10 mg/kg APG pretreatment for 1 h then 10 mg/kg EDF; (F) 10 mg/kg APG pretreatment for 1 h then 25 mg/kg EDF for 14 consecutive days. Oral administration of EDF led to disruption of the intracellular antioxidant machinery which cause the generation of intracellular reactive oxygen species (ROS). However, EDF promotes deleterious effects like oxidative stress, DNA damage, reduced mitochondrial membrane potential, generation of ROS production, activation of caspase 3/9 activities and causing hepato-renal histopathological changes. However, the pretreatment of APG ameliorated the EDF-induced oxidative damage and apoptosis, through their antioxidant activity or by directly scavenging free radical property. Overall, these results suggest that EDF exerts oxidative stress, and APG could be a potent dietary anti-oxidant regimen against EDF-induced toxicity.
Collapse
Affiliation(s)
- Ajaz Ahmad
- Faculty of Life Sciences, Department of Biochemistry, Aligarh Muslim University, 202002, Uttar Pradesh, India
| | - Pooja Kumari
- Faculty of Life Sciences, Department of Zoology, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Masood Ahmad
- Faculty of Life Sciences, Department of Biochemistry, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
16
|
Kumar R, Singh M, Meena J, Singhvi P, Thiyagarajan D, Saneja A, Panda AK. Hyaluronic acid - dihydroartemisinin conjugate: Synthesis, characterization and in vitro evaluation in lung cancer cells. Int J Biol Macromol 2019; 133:495-502. [DOI: 10.1016/j.ijbiomac.2019.04.124] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/06/2019] [Accepted: 04/16/2019] [Indexed: 02/07/2023]
|
17
|
Zhu H, Ji W. Dihydroartemisinin Ameliorated Ovalbumin-Induced Asthma in Mice via Regulation of MiR-183C. Med Sci Monit 2019; 25:3804-3814. [PMID: 31115390 PMCID: PMC6542303 DOI: 10.12659/msm.915399] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The purpose of the present study was to investigate the function and mechanism of dihydroartemisinin (DHA) in treating ovalbumin-induced asthma in BALB/c mice. MATERIAL AND METHODS Thirty female BALB/c mice were randomly separated into 3 groups: the control group, the asthma model group stimulated by ovalbumin (OVA group), and the DHA treatment group (DHA group). The therapeutic effects and potential pharmacological mechanisms of DHA were specifically clarified by examining its effects on asthma-related phenomena, such as body weight, lung function, cell counts in bronchoalveolar lavage fluid (BALF), and hemotoxin and eosin staining. In addition, the expression of inflammatory factors was checked by enzyme-linked immunosorbent assay kits, and fractions of Th17 cells were detected by FACS analysis. Moreover, the downstream molecular pathway of IL-6/Stat3 (interleukin-6/signal transducer and activator of transcription 3) and expression of miR-183C was investigated by western blot and/or quantitative real-time polymerase chain reaction. Luciferase assay was used to reveal the function of miR-183C on the transcriptional regulation of Foxo1 (forkhead box O). RESULTS DHA administration significantly relieved the severity of the asthma through its effect on body weight, survival rate, and airway pressure. DHA was able to ameliorate lung damage in terms of pathological morphology and it reduced the percentage of helper T 17 (Th17) cells and the secretion of cytokines. As a result, the activity of the IL-6/Stat3 pathway was inhibited by DHA. In addition, the adoption of DHA decreased the expression of miR-183C but increased the expression of the transcription factor Foxo1. CONCLUSIONS Our results suggest that the therapeutic effects of DHA on asthma are partially realized via the regulation of miR-183C and IL-6/Stat3 pathway.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Respiratory Disease, Children's Hospital of Soochow University, Suzhou, Jiangsu, China (mainland).,Department of Paediatric, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| | - Wei Ji
- Department of Respiratory Disease, Children's Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| |
Collapse
|
18
|
The induction of apoptosis in BEL-7402 cells by an iridium(III) complex through lysosome–mitochondria pathway. Polyhedron 2018. [DOI: 10.1016/j.poly.2018.09.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
19
|
Guo J, Li Y, Lam CWK, Wang C, Yao M, Zhang W. ZH-1 enhances the anticancer activity of gemcitabine via deoxyribonucleotide synthesis and apoptotic pathway against A549 cells. Food Chem Toxicol 2018; 119:222-230. [PMID: 29653181 DOI: 10.1016/j.fct.2018.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/06/2018] [Accepted: 04/08/2018] [Indexed: 11/18/2022]
Abstract
The purpose of this study was to investigate the inhibitory effect of ZH-1 ((6S,9aS,6aR,9bR)-6-(phenylcarbonyl)-6,6a,9a,9b-tetrahydro-8H-azolidino[3,4-a]b enzo [e]indolizine-7,9-dione) and its potential interaction with gemcitabine in A549 cells. MTT assay showed that the combined use of gemcitabine and ZH-1 presented a significant inhibition effect on A549 cell growth with the cell viability from 82.3 ± 5.6% to 51.0 ± 6.6%. The CI value was 0.60 suggesting a synergistic effect between these two drugs. HPLC-MS/MS data indicated that combined treatment with gemcitabine and ZH-1 induced a significant decrease in deoxyadenosine triphosphate, deoxycytidine triphosphate, deoxyguanosine triphosphate and deoxythymidine triphosphate levels compared with use of gemcitabine alone. Five RNs as well as seven dRNs were considered to be significantly contributive to the discrimination of samples. Furthermore, western blot analysis revealed that the combination treatment caused A549 cell apoptosis via the intrinsic pathway by up-regulating Bax/Bcl-2 ratio, activating caspase-9, caspase-3 and poly-ADP-ribose polymerase, and promoting caspase-7, caspase-9 and poly-ADP-ribose polymerase cleavage. Collectively, the combined treatment with gemcitabine and ZH-1 exerted a strong synergistic action on anticancer activity through growth inhibition, perturbations in ribonucleotides and deoxyribonucleotides and the activation of intrinsic apoptotic signaling pathway.
Collapse
Affiliation(s)
- Jianru Guo
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Yan Li
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Christopher Wai Kei Lam
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Caiyun Wang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Meicun Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China.
| | - Wei Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China.
| |
Collapse
|
20
|
Zhang WY, Yi QY, Wang YJ, Du F, He M, Tang B, Wan D, Liu YJ, Huang HL. Photoinduced anticancer activity studies of iridium(III) complexes targeting mitochondria and tubules. Eur J Med Chem 2018; 151:568-584. [PMID: 29656200 DOI: 10.1016/j.ejmech.2018.04.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/22/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022]
Abstract
Three new iridium (III) complexes [Ir (ppy)2 (ipbc)](PF6) (1), [Ir (bzq)2 (ipbc)](PF6) (2) and [Ir (piq)2 (ipbc)](PF6) (3) were designed and synthesized. All the complexes were tested for anticancer activity using 3-(4,5-dimethylthiazole)-2,5-diphenyltetraazolium bromide (MTT) method. The complexes show no cytotoxic activity toward cancer BEL-7402, SGC-7901, Eca-109, A549, HeLa and HepG2 cells. However, upon irradiation with white light, the complexes display high cytotoxicity against BEL-7402 cells with an IC50 value of 5.5 ± 0.8, 7.3 ± 1.3 and 11.5 ± 1.6 μM for 1, 2 and 3, respectively. AO/EB staining and comet assay show that the complexes can induce apoptosis in BEL-7402 cells. The complexes can increase intracellular ROS and Ca2+ levels and cause a decrease in the mitochondrial membrane potential. Autophagic assays exhibit that the complexes can induce autophagy and regulate the expression of Beclin-1 and LC3 proteins. The cell cycle distribution in BEL-7402 cells was carried out by flow cytometry. The expression of Bcl-2 family proteins was studied by western blot. Additionally, the complexes can release cytochrome c and inhibit the polymerization of α-tubulin. Our study reveals that the complexes inhibit the cell growth in BEL-7402 cells through an ROS-mediated mitochondria dysfunction and targeting tubules pathways. These complexes are a promising new entity for the development of multi-target anticancer drugs.
Collapse
Affiliation(s)
- Wen-Yao Zhang
- Guangdong Engineering & Technology Research Center of Topic Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Qian-Yan Yi
- Guangdong Engineering & Technology Research Center of Topic Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yang-Jie Wang
- Guangdong Engineering & Technology Research Center of Topic Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Fan Du
- Guangdong Engineering & Technology Research Center of Topic Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Miao He
- Guangdong Engineering & Technology Research Center of Topic Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Bing Tang
- Guangdong Engineering & Technology Research Center of Topic Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Dan Wan
- Guangdong Engineering & Technology Research Center of Topic Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yun-Jun Liu
- Guangdong Engineering & Technology Research Center of Topic Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Engineering Research Center for Lead Compounds & Drug Discovery, Guangzhou, 510006, PR China.
| | - Hong-Liang Huang
- School of Life Science and Biopharmaceutical, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
21
|
Zhang B, Zhang Z, Wang J, Yang B, Zhao Y, Rao Z, Gao J. Dihydroartemisinin sensitizes Lewis lung carcinoma cells to carboplatin therapy via p38 mitogen-activated protein kinase activation. Oncol Lett 2018; 15:7531-7536. [PMID: 29740482 PMCID: PMC5934725 DOI: 10.3892/ol.2018.8276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 07/21/2017] [Indexed: 01/18/2023] Open
Abstract
Dihydroartemisinin (DHA), a semi-synthetic derivative of artemisinin isolated from the traditional Chinese herb Artemisia annua, is an effective novel antimalarial agent. Studies have suggested that it also exhibits anticancer effects when administered alone or in combination with conventional chemotherapeutic agents. The present study investigated the therapeutic effect of DHA combined with carboplatin (CBP) on Lewis lung carcinoma (LLC) cells and the possible underlying molecular mechanisms. MTT and clonogenic assays demonstrated that the proliferation activity of LLC cells was inhibited in a dose-dependent manner by DHA combined with CBP. In addition, flow cytometry analysis revealed that cell cycle arrest was induced at the G0/G1 phase and apoptosis was induced following treatment with the combination. When administered in combination with CBP, DHA exhibited more effective anticancer activity compared with DHA or CBP used alone, via increased apoptosis. Following treatment with DHA with or without CBP, the expression of phosphorylated-p38 mitogen-activated protein kinase (MAPK), which can be inhibited with the selective inhibitor SB202190, was detected by western blotting. To summarize, the results of the present study indicated that DHA may sensitize LLC cells to CBP therapy via the activation of p38MAPK, which suggests that a combined treatment of DHA and CBP may be a potential novel therapeutic schedule for lung adenocarcinoma.
Collapse
Affiliation(s)
- Bicheng Zhang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhimin Zhang
- Department of Oncology, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Jun Wang
- Department of Oncology, General Hospital of Jinan Command, People's Liberation Army, Jinan, Shandong 250031, P.R. China
| | - Bo Yang
- Department of Oncology, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Yong Zhao
- Department of Oncology, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Zhiguo Rao
- Department of Oncology, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Jianfei Gao
- Department of Oncology, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
22
|
Jiang J, Geng G, Yu X, Liu H, Gao J, An H, Cai C, Li N, Shen D, Wu X, Zheng L, Mi Y, Yang S. Repurposing the anti-malarial drug dihydroartemisinin suppresses metastasis of non-small-cell lung cancer via inhibiting NF-κB/GLUT1 axis. Oncotarget 2018; 7:87271-87283. [PMID: 27895313 PMCID: PMC5349987 DOI: 10.18632/oncotarget.13536] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 11/02/2016] [Indexed: 12/24/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is an aggressive malignancy and long-term survival remains unsatisfactory for patients with metastatic and recurrent disease. Repurposing the anti-malarial drug dihydroartemisinin (DHA) has been proved to possess potent antitumor effect on various cancers. However, the effects of DHA in preventing the invasion of NSCLC cells have not been studied. In the present study, we determined the inhibitory effects of DHA on invasion and migration and the possible mechanisms involved using A549 and H1975 cells. DHA inhibited in vitro migration and invasion of NSCLC cells even in low concentration with little cytotoxicity. Additionally, low concentration DHA also inhibited Warburg effect in NSCLC cells. Mechanically, DHA negatively regulates NF-κB signaling to inhibit the GLUT1 translocation. Blocking the NF-κB signaling largely abolishes the inhibitory effects of DHA on the translocation of GLUT1 to the plasma membrane and the Warburg effect. Furthermore, GLUT1 knockdown significantly decreased the inhibition of invasion, and migration by DHA. Our results suggested that DHA can inhibit metastasis of NSCLC by targeting glucose metabolism via inhibiting NF-κB signaling pathway and DHA may deserve further investigation in NSCLC treatment.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Thoracic Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Guojun Geng
- Department of Thoracic Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Xiuyi Yu
- Department of Thoracic Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Hongming Liu
- Department of Thoracic Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Jing Gao
- Department of Thoracic Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Hanxiang An
- Department of Medical Oncology, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Chengfu Cai
- Department of Thoracic Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Ning Li
- Department of Thoracic Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Dongyan Shen
- Biobank, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Xiaoqiang Wu
- Biobank, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Lisheng Zheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, People's Republic of China
| | - Yanjun Mi
- Department of Thoracic Surgery, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China.,Department of Medical Oncology, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Shuyu Yang
- Xiamen Diabetes Institution, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| |
Collapse
|
23
|
Tuya N, Wang Y, Tong L, Gao W, Yu R, Xue L. Trichosanthin enhances the antitumor effect of gemcitabine in non-small cell lung cancer via inhibition of the PI3K/AKT pathway. Exp Ther Med 2017; 14:5767-5772. [PMID: 29285119 PMCID: PMC5740788 DOI: 10.3892/etm.2017.5286] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 07/14/2017] [Indexed: 12/30/2022] Open
Abstract
Gemcitabine (GEMZ) is the first-line therapy used against non-small cell lung cancer (NSCLC), and studies have focused on investigating the potential effects of agents combined with GEMZ to enhance the anticancer efficacy in NSCLC. Previous studies have reported that trichosanthin (TCS) has various physiological and pharmacological effects, including anti-human influenza virus enzymes, inhibition of protein synthesis and antitumor activity. The purpose of the present study was to investigate if TCS enhanced the antitumor effects of GEMZ in NSCLC. MTT assay demonstrated that TCS significantly enhanced the cytotoxic effect of GEMZ (P>0.05). Furthermore, a propidium iodide/Αnnexin V staining assay revealed that TCS exerted its pharmacological effect by increasing the apoptotic population. In addition, western blot analysis demonstrated that the combination treatment of TCS with GEMZ further decreased the expression level of phosphoinositide 3-kinase (PI3K) and AKT via regulating the expression of insulin growth factor. The results of the present study demonstrated that TCS enhanced the cytotoxic and apoptotic effects of GEMZ in A549 cells via regulating the PI3K/AKT pathway. In conclusion, these observations may provide a potential rational basis for a combination strategy for chemotherapy treatment of NSCLC.
Collapse
Affiliation(s)
- Naren Tuya
- Department of Biology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Radiation Oncology, General Hospital of Beijing Military Region, Beijing 100700, P.R. China.,Department of Chemotherapy Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia 010020, P.R. China
| | - Yadi Wang
- Department of Biology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Radiation Oncology, General Hospital of Beijing Military Region, Beijing 100700, P.R. China
| | - Lanmei Tong
- Department of Chemotherapy Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia 010020, P.R. China
| | - Weishi Gao
- Department of Chemotherapy Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia 010020, P.R. China
| | - Rong Yu
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital, Hohhot, Inner Mongolia 010020, P.R. China
| | - Liying Xue
- Department of Chemotherapy Oncology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia 010020, P.R. China
| |
Collapse
|
24
|
Poupel F, Aghaei M, Movahedian A, Jafari SM, Shahrestanaki MK. Dihydroartemisinin Induces Apoptosis in Human Bladder Cancer Cell Lines Through Reactive Oxygen Species, Mitochondrial Membrane Potential, and Cytochrome C Pathway. Int J Prev Med 2017; 8:78. [PMID: 29114376 PMCID: PMC5651661 DOI: 10.4103/ijpvm.ijpvm_258_17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/20/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Dihydroartemisinin (DHA) is a semisynthetic derivative of artemisinin and has antiproliferative effect. However, such effects of DHA have not yet been revealed for bladder cancer cells. Methods: We used as bladder cancer cell lines to examine the effect of DHA on the cell viability, cell apoptosis, and monitoring of mitochondrial membrane potential (ΔΨm) changes. Furthermore, the effect of DHA on the reactive oxygen species (ROS) production and cytochrome c release were also detected. We employed MTT assay to investigate the cell proliferation effect of DHA on the EJ-138 and HTB-9 human bladder cancer cells. Annexin/PI staining, caspase-3 activity assay, Bcl-2/Bax protein expression, mitochondrial membrane potential assay, cytochrome c release, and ROS analysis were used for apoptosis detection. Results: DHA significantly reduced cell viability in a dose-dependent manner. Cytotoxicity of DHA was suppressed by N-acetylcysteine. The growth inhibition effect of DHA was related to the induction of cell apoptosis, which were manifested by annexin V-FITC staining, activation of caspase-3. DHA also increased ROS generation, cytochrome c release, and loss of mitochondrial transmembrane potential (ΔΨm) in cells. In addition, the downregulation of regulatory protein Bcl-2 and upregulation of Bax protein by DHA were also observed. Conclusions: These findings demonstrated that DHA induces apoptosis through mitochondrial signaling pathway. These suggest that DHA may be a potential agent for induction of apoptosis in human bladder cancer cells.
Collapse
Affiliation(s)
- Farhad Poupel
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmoud Aghaei
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Movahedian
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyyed Mehdi Jafari
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Keyvanloo Shahrestanaki
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
25
|
Chen H, Gu S, Dai H, Li X, Zhang Z. Dihydroartemisinin Sensitizes Human Lung Adenocarcinoma A549 Cells to Arsenic Trioxide via Apoptosis. Biol Trace Elem Res 2017; 179:203-212. [PMID: 28261759 DOI: 10.1007/s12011-017-0975-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/15/2017] [Indexed: 11/29/2022]
Abstract
Recent studies have shown that arsenic trioxide (ATO) is an effective anti-cancer drug for treatment of acute promyelocytic leukemia and other types of human cancer. However, we have found that lung cancer cells constantly develop a high level of resistance to ATO. In this study, we have explored a possibility of combination of dihydroartemisinin (DHA) and ATO treatments to reduce ATO resistance of lung cancer cells. We determined the combinatory effects of DHA and ATO on cytotoxicity of human lung adenocarcinoma (A549) cells. We showed that co-exposure to DHA and ATO of A549 cells synergistically increased the cytotoxicity and apoptotic cell death in the cells. We found that the synergistic effect of DHA and ATO in promoting apoptosis mainly resulted from increased cellular level of reactive oxygen species (ROS) and DNA damage. ATO alone only exerted moderate growth inhibitory effects on A549 cells. The results indicate that DHA can significantly sensitize ATO-induced cytotoxicity of A549 lung cancer cells through apoptosis mediated by ROS-induced DNA damage. Interestingly, we found that the combinatory treatment of DHA and ATO did not result in significant adverse effects in normal human bronchial epithelial (HBE) cells. Our results further provide evidence for the potential application of combinatory effects of DHA and ATO as a safe therapy for human lung cancer.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Shiyan Gu
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Huangmei Dai
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xinyang Li
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zunzhen Zhang
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
26
|
Cancer combination therapies with artemisinin-type drugs. Biochem Pharmacol 2017; 139:56-70. [DOI: 10.1016/j.bcp.2017.03.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/28/2017] [Indexed: 01/28/2023]
|
27
|
Tang B, Wan D, Lai SH, Yang HH, Zhang C, Wang XZ, Zeng CC, Liu YJ. Design, synthesis and evaluation of anticancer activity of ruthenium (II) polypyridyl complexes. J Inorg Biochem 2017; 173:93-104. [DOI: 10.1016/j.jinorgbio.2017.04.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 10/19/2022]
|
28
|
Jin H, Jiang AY, Wang H, Cao Y, Wu Y, Jiang XF. Dihydroartemisinin and gefitinib synergistically inhibit NSCLC cell growth and promote apoptosis via the Akt/mTOR/STAT3 pathway. Mol Med Rep 2017; 16:3475-3481. [PMID: 28713965 DOI: 10.3892/mmr.2017.6989] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 02/20/2017] [Indexed: 11/06/2022] Open
Abstract
Non‑small cell lung cancer (NSCLC) is among the leading causes of cancer‑associated mortality worldwide. In clinical practice, therapeutic strategies based on drug combinations are often used for the treatment of various types of cancer. The present study aimed to investigate the effects of the combination of dihydroartemisinin (DHA) and gefitinib on NSCLC. Cell Counting kit 8 assay was used to evaluate cell viability. Transwell assays were performed to investigate cellular migration and invasion, and cellular apoptosis was evaluated using the terminal deoxynucleotidyl transferase dUTP nick‑end labeling assay. Flow cytometry was used to investigate cell cycle distribution and the expression levels of target proteins were determined using western blot analysis. The results of the present study demonstrated that DHA (5, 10, 20, 50 and 100 µM) reduced cancer cell viability in a dose‑dependent manner in the NCI‑H1975 human NSCLC cell line and significantly enhanced gefitinib‑induced apoptosis. Furthermore, DHA and gefitinib co‑administration induced cell cycle arrest in G2/M phase, which was associated with a marked decline in the protein expression levels of G2/M regulatory proteins, including cyclin B1 and cyclin‑dependent kinase 1. The addition of DHA appeared to potentiate the inhibitory actions of gefitinib on the migratory and invasive capabilities of NCI‑H1975 cells. DHA and gefitinib co‑administration also downregulated the expression levels of phosphorylated (p)‑Akt, p‑mechanistic target of rapamycin, p‑signal transducer and activator of transcription 3 and B‑cell lymphoma 2 (Bcl‑2), and upregulated the expression of Bcl‑2‑associated X protein. In conclusion, the present results suggested that the combination of DHA and gefitinib may have potential as a novel and more effective therapeutic strategy for the treatment of patients with NSCLC.
Collapse
Affiliation(s)
- Hong Jin
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ai-Ying Jiang
- Department of Pneumology, Hongqi Hospital, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Han Wang
- Department of Clinical Skills Center, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yong Cao
- Department of Pathology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yan Wu
- Department of Medical Research Center, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Xiao-Feng Jiang
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
29
|
Zhang Y, Liang Y, He C. Anticancer activities and mechanisms of heat-clearing and detoxicating traditional Chinese herbal medicine. Chin Med 2017; 12:20. [PMID: 28702078 PMCID: PMC5506596 DOI: 10.1186/s13020-017-0140-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023] Open
Abstract
In traditional Chinese medicine (TCM) theory, pathogenic heat and toxins, which are akin to the inflammatory factors, are the causes of cancer and could promote its virulent development. Therefore, heat-clearing and detoxicating (HCD) herbs are essential components of TCM formulas for cancer treatment. An increasing interest has been focused on the study of HCD herbs and accumulated evidences have shown that HCD herbs or HCD herbs-based formulas exhibited remarkable anticancer effects when used alone or combined with other therapeutic approaches. Some of the HCD herb-derived products have been tested in clinical trials. Studies revealed that extracts or pure compounds of the HCD herbs showed a broad anticancer spectrum against both solid and hematologic malignancies without significant toxic effects. Notably, some HCD herbs or formulas could strongly enhance the anticancer activities of chemo- or radio-therapy and alleviate their side effects. The anticancer activities of HCD herb exacts or the pure compounds were reported to be through multiple cellular or molecular mechanisms, such as induction of cancer cell apoptosis, differentiation and cell cycle arrest, inhibition of cancer cell growth, invasion and metastasis, and inhibition of tumor angiogenesis. In this review, we provide comprehensive analysis and summary of research progress and future prospects in this field to facilitate the further study and application of HCD herbs.
Collapse
Affiliation(s)
- Yulin Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, N22-7038, Avenida da Universidade, Taipa, Macao, 999078 China
| | - Yeer Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, N22-7038, Avenida da Universidade, Taipa, Macao, 999078 China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, N22-7038, Avenida da Universidade, Taipa, Macao, 999078 China
| |
Collapse
|
30
|
Wong YK, Xu C, Kalesh KA, He Y, Lin Q, Wong WSF, Shen HM, Wang J. Artemisinin as an anticancer drug: Recent advances in target profiling and mechanisms of action. Med Res Rev 2017. [PMID: 28643446 DOI: 10.1002/med.21446] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Artemisinin and its derivatives (collectively termed as artemisinins) are among the most important and effective antimalarial drugs, with proven safety and efficacy in clinical use. Beyond their antimalarial effects, artemisinins have also been shown to possess selective anticancer properties, demonstrating cytotoxic effects against a wide range of cancer types both in vitro and in vivo. These effects appear to be mediated by artemisinin-induced changes in multiple signaling pathways, interfering simultaneously with multiple hallmarks of cancer. Great strides have been taken to characterize these pathways and to reveal their anticancer mechanisms of action of artemisinin. Moreover, encouraging data have also been obtained from a limited number of clinical trials to support their anticancer property. However, there are several key gaps in knowledge that continue to serve as significant barriers to the repurposing of artemisinins as effective anticancer agents. This review focuses on important and emerging aspects of this field, highlighting breakthroughs in unresolved questions as well as novel techniques and approaches that have been taken in recent studies. We discuss the mechanism of artemisinin activation in cancer, novel and significant findings with regards to artemisinin target proteins and pathways, new understandings in artemisinin-induced cell death mechanisms, as well as the practical issues of repurposing artemisinin. We believe these will be important topics in realizing the potential of artemisinin and its derivatives as safe and potent anticancer agents.
Collapse
Affiliation(s)
- Yin Kwan Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chengchao Xu
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Karunakaran A Kalesh
- Department of Chemical Engineering, Imperial College London, London, United Kingdom
| | - Yingke He
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigang Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
31
|
Artemisinin and its derivatives in cancer therapy: status of progress, mechanism of action, and future perspectives. Cancer Chemother Pharmacol 2017; 79:451-466. [DOI: 10.1007/s00280-017-3251-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/03/2017] [Indexed: 12/21/2022]
|
32
|
Farnesylthiosalicylic acid sensitizes hepatocarcinoma cells to artemisinin derivatives. PLoS One 2017; 12:e0171840. [PMID: 28182780 PMCID: PMC5300221 DOI: 10.1371/journal.pone.0171840] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/26/2017] [Indexed: 01/03/2023] Open
Abstract
Dihydroartemisinin (DHA) and artesunate (ARS), two artemisinin derivatives, have efficacious anticancer activities against human hepatocarcinoma (HCC) cells. This study aims to study the anticancer action of the combination treatment of DHA/ARS and farnesylthiosalicylic acid (FTS), a Ras inhibitor, in HCC cells (Huh-7 and HepG2 cell lines). FTS pretreatment significantly enhanced DHA/ARS-induced phosphatidylserine (PS) externalization, Bak/Bax activation, mitochondrial membrane depolarization, cytochrome c release, and caspase-8 and -9 activations, characteristics of the extrinsic and intrinsic apoptosis. Pretreatment with Z-IETD-FMK (caspase-8 inhibitor) potently prevented the cytotoxicity of the combination treatment of DHA/ARS and FTS, and pretreatment with Z-VAD-FMK (pan-caspase inhibitor) significantly inhibited the loss of ΔΨm induced by DHA/ARS treatment or the combination treatment of DHA/ARS and FTS in HCC cells. Furthermore, silencing Bak/Bax modestly but significantly inhibited the cytotoxicity of the combination treatment of DHA/ARS and FTS. Interestingly, pretreatment with an antioxidant N-Acetyle-Cysteine (NAC) significantly prevented the cytotoxicity of the combination treatment of DHA and FTS instead of the combination treatment of ARS and FTS, suggesting that reactive oxygen species (ROS) played a key role in the anticancer action of the combination treatment of DHA and FTS. Similar to FTS, DHA/ARS also significantly prevented Ras activation. Collectively, our data demonstrate that FTS potently sensitizes Huh-7 and HepG2 cells to artemisinin derivatives via accelerating the extrinsic and intrinsic apoptotic pathways.
Collapse
|
33
|
Xu B, Tao ZZ. Piceatannol Enhances the Antitumor Efficacy of Gemcitabine in Human A549 Non-Small Cell Lung Cancer Cells. Oncol Res 2016; 22:213-217. [PMID: 26351210 PMCID: PMC7838451 DOI: 10.3727/096504015x14386062091398] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
To enhance the anticancer efficacy of gemcitabine in the treatment of non-small cell lung cancer (NSCLC), the potential synergistic effect of piceatannol on gemcitabine cytotoxicity was investigated in the human NSCLC A459 cell line. The MTT cell viability assay showed that piceatannol significantly enhanced the cytotoxic effects of gemcitabine by lowering the gemcitabine IC50 value. Flow cytometry analysis revealed that piceatannol exerted its pharmacological effect mainly by increasing the late apoptotic population. Western blot analysis showed that gemcitabine induced the expression of the proapoptotic proteins Bad and Bak, and pretreatment with piceatannol further increased Bak expression, leading to an increased number of cells undergoing late apoptosis. The findings from this study show that piceatannol can enhance the cytotoxic effects of gemcitabine by enhancing expression of the proapoptotic protein Bak, thereby providing the rational basis for a novel combination strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Bin Xu
- Department of Oncology, Wuhan University Renmin Hospital, Wuhan, Hubei, P.R. China
| | | |
Collapse
|
34
|
Qin G, Zhao C, Zhang L, Liu H, Quan Y, Chai L, Wu S, Wang X, Chen T. Dihydroartemisinin induces apoptosis preferentially via a Bim-mediated intrinsic pathway in hepatocarcinoma cells. Apoptosis 2016; 20:1072-86. [PMID: 25935454 DOI: 10.1007/s10495-015-1132-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This report is designed to dissect the detail molecular mechanism by which dihydroartemisinin (DHA), a derivative of artemisinin, induces apoptosis in human hepatocellular carcinoma (HCC) cells. DHA induced a loss of the mitochondrial transmemberane potential (ΔΨm), release of cytochrome c, activation of caspases, and externalization of phosphatidylserine indicative of apoptosis induction. Compared with the modest inhibitory effects of silencing Bax, silencing Bak largely prevented DHA-induced ΔΨm collapse and apoptosis though DHA induced a commensurable activation of Bax and Bak, demonstrating a key role of the Bak-mediated intrinsic apoptosis pathway. DHA did not induce Bid cleavage and translocation from cytoplasm to mitochondria and had little effects on the expressions of Puma and Noxa, but did increase Bim and Bak expressions and decrease Mcl-1 expression. Furthermore, the cytotoxicity of DHA was remarkably reduced by silencing Bim, and modestly but significantly reduced by silencing Puma or Noxa. Silencing Bim or Noxa preferentially reduced DHA-induced Bak activation, while silencing Puma preferentially reduced DHA-induced Bax activation, demonstrating that Bim and to a lesser extent Noxa act as upstream mediators to trigger the Bak-mediated intrinsic apoptosis pathway. In addition, silencing Mcl-1 enhanced DHA-induced Bak activation and apoptosis. Taken together, our data demonstrate a crucial role of Bim in preferentially regulating the Bak/Mcl-1 rheostat to mediate DHA-induced apoptosis in HCC cells.
Collapse
Affiliation(s)
- Guiqi Qin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Qin G, Wu L, Liu H, Pang Y, Zhao C, Wu S, Wang X, Chen T. Artesunate induces apoptosis via a ROS-independent and Bax-mediated intrinsic pathway in HepG2 cells. Exp Cell Res 2015; 336:308-17. [PMID: 26163896 DOI: 10.1016/j.yexcr.2015.07.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 06/13/2015] [Accepted: 07/06/2015] [Indexed: 01/06/2023]
Abstract
This study aims to explore the detail molecular mechanism by which artesunate (ARS), an artemisinin derivative, induces apoptosis in HepG2 cells. ARS induced a loss of mitochondrial transmemberane potential (ΔΨm), phosphatidylserine (PS) externalization, as well as activations of Bax/Bak and caspases indicative of apoptosis induction. Silencing Bax but not Bak significantly inhibited ARS-induced apoptosis, demonstrating the key role of the Bax-mediated intrinsic pathway. Although ARS increased intracellular reactive oxygen species (ROS), ARS-induced apoptosis was neither prevented by pretreatment with ROS scavengers nor potentiated by pretreatment with l-buthionine-sulfoximine (BSO) that enhanced the ARS-induced intracellular ROS generation, demonstrating that ROS was not involved in ARS-induced apoptosis. In addition, ARS did not induce Bid translocation to mitochondria, and the cytotoxicity of ARS was not prevented by silencing Bim, Puma or Mcl-1, but was significantly enhanced by HA14-1 pretreatment, demonstrating that Bcl-2/-xl instead of Bid and Bim as well as Puma may be the upstream factor to regulate the Bax-mediated intrinsic pathway. Collectively, our data demonstrate that ARS induces ROS-independent apoptosis via the Bax-mediated intrinsic pathway in HepG2 cells.
Collapse
Affiliation(s)
- Guiqi Qin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, PR China
| | - Liping Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, PR China
| | - Hongyu Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, PR China
| | - Yilin Pang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, PR China
| | - Chubiao Zhao
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, PR China
| | - Shengnan Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, PR China
| | - Xiaoping Wang
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, PR China.
| | - Tongsheng Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, PR China.
| |
Collapse
|
36
|
Li Y, Rouhi O, Chen H, Ramirez R, Borgia JA, Deng Y. RNA-Seq and Network Analysis Revealed Interacting Pathways in TGF-β-Treated Lung Cancer Cell Lines. Cancer Inform 2015; 13:129-40. [PMID: 25991908 PMCID: PMC4384765 DOI: 10.4137/cin.s14073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/02/2014] [Accepted: 11/08/2014] [Indexed: 01/15/2023] Open
Abstract
Whole transcriptome shotgun sequencing (RNA-Seq) is a useful tool for analyzing the transcriptome of a biological sample. With appropriate statistical and bioinformatic processing, this platform is capable of identifying significant differences in gene expression within the transcriptome and permits pathway and network analyses to determine how these genes interact biologically. In this study, we examined gene expression in two lung adenocarcinoma cell lines (H358 and A459) that were treated with transforming growth factor-β (TGF-β) as a model for induction of the epithelial-to-mesenchymal transition (EMT), commonly associated with disease progression. We performed this study in order to illustrate a workflow for identifying interesting genes and processes that are regulated early in EMT and to determine their gene pathway/network relationships and regulation. With this, we identified 137 upregulated and 32 downregulated genes common to both cell lines after TGF-β treatment that represent components of multiple canonical pathways and biological networks associated with the induction of EMT. These findings were also verified against reposited Affymetrix U133a expression profiles from multiple trials examining metastatic progression in patient cohorts (n = 731 total) to further establish the clinical relevance and translational significance of the model system. Together, these findings help validate the relevance of the TGF-β model for the study of EMT and provide new insights into early events in EMT.
Collapse
Affiliation(s)
- Yan Li
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Omid Rouhi
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Hankui Chen
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Rolando Ramirez
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Jeffrey A Borgia
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA. ; Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Youping Deng
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA. ; Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
37
|
Mi YJ, Geng GJ, Zou ZZ, Gao J, Luo XY, Liu Y, Li N, Li CL, Chen YQ, Yu XY, Jiang J. Dihydroartemisinin inhibits glucose uptake and cooperates with glycolysis inhibitor to induce apoptosis in non-small cell lung carcinoma cells. PLoS One 2015; 10:e0120426. [PMID: 25799586 PMCID: PMC4370589 DOI: 10.1371/journal.pone.0120426] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/22/2015] [Indexed: 01/16/2023] Open
Abstract
Despite recent advances in the therapy of non-small cell lung cancer (NSCLC), the chemotherapy efficacy against NSCLC is still unsatisfactory. Previous studies show the herbal antimalarial drug dihydroartemisinin (DHA) displays cytotoxic to multiple human tumors. Here, we showed that DHA decreased cell viability and colony formation, induced apoptosis in A549 and PC-9 cells. Additionally, we first revealed DHA inhibited glucose uptake in NSCLC cells. Moreover, glycolytic metabolism was attenuated by DHA, including inhibition of ATP and lactate production. Consequently, we demonstrated that the phosphorylated forms of both S6 ribosomal protein and mechanistic target of rapamycin (mTOR), and GLUT1 levels were abrogated by DHA treatment in NSCLC cells. Furthermore, the upregulation of mTOR activation by high expressed Rheb increased the level of glycolytic metabolism and cell viability inhibited by DHA. These results suggested that DHA-suppressed glycolytic metabolism might be associated with mTOR activation and GLUT1 expression. Besides, we showed GLUT1 overexpression significantly attenuated DHA-triggered NSCLC cells apoptosis. Notably, DHA synergized with 2-Deoxy-D-glucose (2DG, a glycolysis inhibitor) to reduce cell viability and increase cell apoptosis in A549 and PC-9 cells. However, the combination of the two compounds displayed minimal toxicity to WI-38 cells, a normal lung fibroblast cell line. More importantly, 2DG synergistically potentiated DHA-induced activation of caspase-9, -8 and -3, as well as the levels of both cytochrome c and AIF of cytoplasm. However, 2DG failed to increase the reactive oxygen species (ROS) levels elicited by DHA. Overall, the data shown above indicated DHA plus 2DG induced apoptosis was involved in both extrinsic and intrinsic apoptosis pathways in NSCLC cells.
Collapse
Affiliation(s)
- Yan-jun Mi
- Department of thoracic surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Department of Medical Oncology, Chenggong Hospital of Xiamen University, Xiamen, China
| | - Guo-jun Geng
- Department of thoracic surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zheng-zhi Zou
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Jing Gao
- Department of Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xian-yang Luo
- Department of Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yu Liu
- Department of thoracic surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ning Li
- Department of thoracic surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chun-lei Li
- Department of thoracic surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yu-qiang Chen
- Department of Medical Oncology, Chenggong Hospital of Xiamen University, Xiamen, China
| | - Xiu-yi Yu
- Department of thoracic surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
- * E-mail: (XYY); (JJ)
| | - Jie Jiang
- Department of thoracic surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
- * E-mail: (XYY); (JJ)
| |
Collapse
|
38
|
Zhao C, Qin G, Gao W, Chen J, Liu H, Xi G, Li T, Wu S, Chen T. Potent proapoptotic actions of dihydroartemisinin in gemcitabine-resistant A549 cells. Cell Signal 2014; 26:2223-33. [PMID: 25018064 DOI: 10.1016/j.cellsig.2014.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 06/16/2014] [Accepted: 07/04/2014] [Indexed: 11/17/2022]
Abstract
Our recent studies have demonstrated the key roles of reactive oxygen species (ROS)-mediated caspase-8- and Bax-dependent apoptotic pathways in dihydroartemisinin (DHA)-induced apoptosis of A549 cells. This report is designed to investigate the proapoptotic mechanisms of DHA in gemcitabine (Gem)-resistant A549 (A549GR) cells. A549GR cells exhibited lower basal antioxidant capacity, higher level of basal ROS and intracellular Fe(2+) than Gem-sensitive A549 (A549) cells. In contrast to the sluggish ROS generation induced by Gem, DHA induced a rapid ROS generation within 30min. Moreover, Gem induced similar ROS generation in both cell lines, while DHA induced more ROS generation in A549GR cells than in A549 cells. More importantly, after treatment with DHA, A549GR cells showed more potent induction in Bax activation, loss of mitochondrial membrane potential (ΔΨm), caspase activation and apoptosis than A549 cells. Furthermore, NAC pretreatment potently prevented DHA-induced ROS generation and loss of ΔΨm as well as apoptosis, and silencing Bax by shRNA or inhibition of one of caspase-3, -8 and -9 also significantly prevented DHA-induced apoptosis in both cell lines, indicating the key roles of ROS and Bax as well as the caspases. Collectively, DHA presents more potent proapoptotic actions in A549GR cells preferentially over normal A549 cells via ROS-dependent apoptotic pathway, in which Bax and caspases are involved.
Collapse
Affiliation(s)
- Chubiao Zhao
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Guiqi Qin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Weijie Gao
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jingqin Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Hongyu Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Gaina Xi
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Tan Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Shengnan Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China
| | - Tongsheng Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Life Science, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
39
|
Chen J, Liu H, Zhao C, Qin G, Xi G, Li T, Wang X, Chen T. One-step reduction and PEGylation of graphene oxide for photothermally controlled drug delivery. Biomaterials 2014; 35:4986-95. [DOI: 10.1016/j.biomaterials.2014.02.032] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/20/2014] [Indexed: 01/09/2023]
|