1
|
Vafaeinik F, Zhang L, Lee YJ. Low extracellular pH enhances TRAIL-induced apoptosis by downregulating Mcl-1 expression. Exp Cell Res 2025; 447:114481. [PMID: 40024506 DOI: 10.1016/j.yexcr.2025.114481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
We previously reported that low extracellular pH promotes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through the mitochondria-mediated caspase signal transduction pathway. In this study, we further investigated the mechanism of low extracellular pH on TRAIL-induced apoptosis. When human colorectal carcinoma HCT116 cells were treated with TRAIL for 4 h, significant cytotoxicity was observed at pH 6.3, while cytotoxic effects were notably reduced at pH 7.2. These findings suggest that TRAIL's cytotoxic effects on human colorectal cancer cells are enhanced in low pH environments following TRAIL treatment. Similar results were observed in human pancreatic adenocarcinoma BxPC-3 cells. Interestingly, TRAIL was found to downregulate the levels of anti-apoptotic proteins, such as Mcl-1. This was confirmed by the knock-in (KI) of an Mcl-1 phosphorylation site mutant in HCT116 cells, which blocked TRAIL-induced Mcl-1 downregulation and the subsequent apoptotic response. These results indicate that Mcl-1 mediates TRAIL resistance in the Mcl-1 KI cells. Additionally, our results revealed that TRAIL significantly induced JNK phosphorylation in HCT116 cells, suggesting the involvement of JNK in TRAIL-induced cell death in colorectal cancer cells. Our findings demonstrate that low extracellular pH enhances TRAIL-induced cytotoxicity, particularly at pH 6.3 and 6.6. Moreover, the anti-apoptotic Bcl-2 family member Mcl-1 is an important target of TRAIL in colorectal carcinoma HCT116 cells under different low pH conditions. TRAIL triggered a rapid decline in Mcl-1, suggesting that Mcl-1 downregulation is crucial for TRAIL-induced apoptosis.
Collapse
Affiliation(s)
- Farzaneh Vafaeinik
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Lin Zhang
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90007, USA; Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90007, USA
| | - Yong J Lee
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
2
|
Zinnah KMA, Munna AN, Park SY. Optimizing autophagy modulation for enhanced TRAIL-mediated therapy: Unveiling the superiority of late-stage inhibition over early-stage inhibition to overcome therapy resistance in cancer. Basic Clin Pharmacol Toxicol 2025; 136:e14110. [PMID: 39668304 DOI: 10.1111/bcpt.14110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/13/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024]
Abstract
Autophagy is a vital mechanism that eliminates large cytoplasmic components via lysosomal degradation to maintain cellular homeostasis. The role of autophagy in cancer treatment has been studied extensively. Autophagy primarily prevents tumour initiation by maintaining genomic stability and preventing cellular inflammation. However, autophagy also supports cancer cell survival and growth by providing essential nutrients for therapeutic resistance. Thus, autophagy has emerged as a promising strategy for overcoming resistance and enhancing anti-cancer therapy. Inhibiting autophagy significantly improves the sensitivity of lung, colorectal, breast, liver and prostate cancer cells to tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). This review investigates the intricate interplay between autophagy modulation and TRAIL-based therapy, specifically focussing on comparing the efficacy of late-stage autophagy inhibition versus early-stage inhibition in overcoming cancer resistance. We expose the distinctive advantages of late-stage autophagy inhibition by exploring the mechanisms underlying autophagy's impact on TRAIL sensitivity. Current preclinical and clinical investigations are inspected, showing the potential of targeting late-stage autophagy for sensitizing resistant cancer cells to TRAIL-induced apoptosis. This review emphasizes the significance of optimizing autophagy modulation to enhance TRAIL-mediated therapy and overcome the challenge of treatment resistance in cancer. We offer insights and recommendations for guiding the development of potential therapeutic strategies aimed at overcoming the challenges posed by treatment-resistant cancers.
Collapse
Affiliation(s)
- Kazi Mohammad Ali Zinnah
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
- Faculty of Biotechnology and Genetic Engineering, Department of Animal and Fish Biotechnology, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Ali Newaz Munna
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| |
Collapse
|
3
|
Liu Y, Meng Y, Zhang J, Gu L, Shen S, Zhu Y, Wang J. Pharmacology Progresses and Applications of Chloroquine in Cancer Therapy. Int J Nanomedicine 2024; 19:6777-6809. [PMID: 38983131 PMCID: PMC11232884 DOI: 10.2147/ijn.s458910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/07/2024] [Indexed: 07/11/2024] Open
Abstract
Chloroquine is a common antimalarial drug and is listed in the World Health Organization Standard List of Essential Medicines because of its safety, low cost and ease of use. Besides its antimalarial property, chloroquine also was used in anti-inflammatory and antivirus, especially in antitumor therapy. A mount of data showed that chloroquine mainly relied on autophagy inhibition to exert its antitumor effects. However, recently, more and more researches have revealed that chloroquine acts through other mechanisms that are autophagy-independent. Nevertheless, the current reviews lacked a comprehensive summary of the antitumor mechanism and combined pharmacotherapy of chloroquine. So here we focused on the antitumor properties of chloroquine, summarized the pharmacological mechanisms of antitumor progression of chloroquine dependent or independent of autophagy inhibition. Moreover, we also discussed the side effects and possible application developments of chloroquine. This review provided a more systematic and cutting-edge knowledge involved in the anti-tumor mechanisms and combined pharmacotherapy of chloroquine in hope of carrying out more in-depth exploration of chloroquine and obtaining more clinical applications.
Collapse
Affiliation(s)
- Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Liwei Gu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Shengnan Shen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
- Department of Pharmacological Sciences, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| |
Collapse
|
4
|
Pavić K, Poje G, Carvalho LPD, Held J, Rajić Z. Synthesis, antiproliferative and antiplasmodial evaluation of new chloroquine and mefloquine-based harmiquins. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:537-558. [PMID: 38147482 DOI: 10.2478/acph-2023-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/09/2023] [Indexed: 12/28/2023]
Abstract
Here we present the synthesis and evaluation of the biological activity of new hybrid compounds, ureido-type (UT) harmiquins, based on chloroquine (CQ) or mefloquine (MQ) scaffolds and β-carboline alkaloid harmine against cancer cell lines and Plasmodium falciparum. The hybrids were prepared from the corresponding amines by 1,1'-carbonyldiimidazole (CDI)-mediated synthesis. In vitro evaluation of the biological activity of the title compounds revealed two hit compounds. Testing of the antiproliferative activity of the new UT harmiquins, and previously prepared triazole-(TT) and amide-type (AT) CQ-based harmiquins, against a panel of human cell lines, revealed TT harmiquine 16 as the most promising compound, as it showed pronounced and selective activity against the tumor cell line HepG2 (IC 50 = 5.48 ± 3.35 μmol L-1). Screening of the antiplasmodial activities of UT harmiquins against erythrocytic stages of the Plasmodium life cycle identified CQ-based UT harmiquine 12 as a novel antiplasmodial hit because it displayed low IC 50 values in the submicromolar range against CQ-sensitive and resistant strains (IC 50 0.06 ± 0.01, and 0.19 ± 0.02 μmol L-1, respectively), and exhibited high selectivity against Plasmodium, compared to mammalian cells (SI = 92).
Collapse
Affiliation(s)
- Kristina Pavić
- 1University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medicinal Chemistry 10 000 Zagreb, Croatia
| | - Goran Poje
- 1University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medicinal Chemistry 10 000 Zagreb, Croatia
| | | | - Jana Held
- 2University of Tübingen, Institute of Tropical Medicine, 72074, Tübingen Germany
- 3German Center for Infection Research (DZIF), 72074, Tübingen Germany
| | - Zrinka Rajić
- 1University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medicinal Chemistry 10 000 Zagreb, Croatia
| |
Collapse
|
5
|
Khan R, Panja S, Ding L, Tang S, Tang W, Kapoor E, Bennett RG, Oupický D. Polymeric Chloroquine as an Effective Antimigration Agent in the Treatment of Pancreatic Cancer. Mol Pharm 2022; 19:4631-4643. [PMID: 36346968 DOI: 10.1021/acs.molpharmaceut.2c00596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hydroxychloroquine (HCQ) has been the subject of multiple recent preclinical and clinical studies for its beneficial use in the combination treatments of different types of cancers. Polymeric HCQ (PCQ), a macromolecular multivalent version of HCQ, has been shown to be effective in various cancer models both in vitro and in vivo as an inhibitor of cancer cell migration and experimental lung metastasis. Here, we present detailed in vitro studies that show that low concentrations of PCQ can efficiently inhibit cancer cell migration and colony formation orders of magnitude more effectively compared to HCQ. After intraperitoneal administration of PCQ in vivo, high levels of tumor accumulation and penetration are observed, combined with strong antimetastatic activity in an orthotopic pancreatic cancer model. These studies support the idea that PCQ may be effectively used at low doses as an adjuvant in the therapy of pancreatic cancer. In conjunction with previously published literature, these studies further undergird the potential of PCQ as an anticancer agent.
Collapse
Affiliation(s)
- Rubayat Khan
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Siyuan Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Ekta Kapoor
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Robert G Bennett
- Department of Internal Medicine and Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Research Service, Nebraska-Western Iowa Health Care System, Omaha, Nebraska 68105, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
6
|
Novel 7-Chloro-(4-thioalkylquinoline) Derivatives: Synthesis and Antiproliferative Activity through Inducing Apoptosis and DNA/RNA Damage. Pharmaceuticals (Basel) 2022; 15:ph15101234. [PMID: 36297346 PMCID: PMC9607427 DOI: 10.3390/ph15101234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/29/2022] Open
Abstract
A series of 78 synthetic 7-chloro-(4-thioalkylquinoline) derivatives were investigated for cytotoxic activity against eight human cancer as well as 4 non-tumor cell lines. The results showed, with some exceptions, that sulfanyl 5-40 and sulfinyl 41-62 derivatives exhibited lower cytotoxicity for cancer cell lines than those of well-described sulfonyl N-oxide derivatives 63-82. As for compound 81, the most pronounced selectivity (compared against BJ and MRC-5 cells) was observed for human cancer cells from HCT116 (human colorectal cancer with wild-type p53) and HCT116p53-/- (human colorectal cancer with deleted p53), as well as leukemia cell lines (CCRF-CEM, CEM-DNR, K562, and K562-TAX), lung (A549), and osteosarcoma cells (U2OS). A good selectivity was also detected for compounds 73 and 74 for leukemic and colorectal (with and without p53 deletion) cancer cells (compared to MRC-5). At higher concentrations (5 × IC50) against the CCRF-CEM cancer cell line, we observe the accumulation of the cells in the G0/G1 cell phase, inhibition of DNA and RNA synthesis, and induction of apoptosis. In addition, X-ray data for compound 15 is being reported. These results provide useful scientific data for the development of 4-thioalkylquinoline derivatives as a new class of anticancer candidates.
Collapse
|
7
|
Gamea GA, Elmehy DA, Salama AM, Soliman NA, Afifi OK, Elkaliny HH, Abo El gheit RE, El-Ebiary AA, Tahoon DM, Elkholy RA, Shoeib SM, Eleryan MA, Younis SS. Direct and indirect antiparasitic effects of chloroquine against the virulent RH strain of Toxoplasma gondii: An experimental study. Acta Trop 2022; 232:106508. [PMID: 35568067 DOI: 10.1016/j.actatropica.2022.106508] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND Toxoplasmosis is a deleterious parasitic disease with harmful impact on both humans and animals. The present study was carried out to evaluate the antiparasitic effect of chloroquine (CQ), spiramycin (SP), and combination of both against the highly virulent RH HXGPRT (-) strain of Toxoplasma gondii (T. gondii) and to explore the mechanisms underlying such effect. METHODS We counted the tachyzoites in the peritoneal fluid and liver smears of mice and performed scanning and transmission electron microscopy and immunofluorescence staining of tachyzoites. Moreover, relative caspase 3 gene expression was measured by real time polymerase chain reaction of liver tissues and immunoassay of anti-apoptotic markers [B cell lymphoma-2 (Bcl-2) and X-chromosome linked inhibitor of apoptosis (XIAP)] and interferon gamma (IFN-γ) was done in liver tissues by ELISA. In addition, we estimated serum levels of aspartate transaminase (AST) and alanine transaminase (ALT) and performed histopathological examination of liver sections for scoring of inflammation. RESULTS We found that both CQ and CQ/SP combination significantly reduced parasitic load in the peritoneal fluid and liver smears, induced apical disruption of tachyzoites, triggered host cell apoptosis through elevation of relative caspase 3 gene expression and suppression of both Bcl-2 and XIAP. Also, they upregulated IFN-γ level, reduced serum AST and ALT, and ameliorated liver inflammation. CONCLUSIONS Either of CQ and CQ/SP combination was more effective than SP alone against T. gondii with the CQ/SP combination being more efficient. Therefore, adding CQ to other anti-Toxoplasma therapeutic regimens may be considered in future research.
Collapse
|
8
|
Memari F, Mirzavi F, Jalili-Nik M, Afshari AR, Ghorbani A, Soukhtanloo M. Tumor-Inhibitory Effects of Zerumbone Against HT-29 Human Colorectal Cancer Cells. Int J Toxicol 2022; 41:402-411. [PMID: 35719111 DOI: 10.1177/10915818221104417] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Colorectal cancer (CRC) is the second cause of cancer-associated death globally. Recently, herbal medicinal products and, in particular, zerumbone have been widely studied and used for cancer treatment as they induce significant anti-cancer effects. However, there is limited information about the anti-cancer effects of zerumbone in CRC. Therefore, we aimed to investigate the in vitro anti-cancer effects of the zerumbone in CRC, focusing on cell apoptosis and migration. Anti-proliferative and anti-migratory effects of zerumbone on HT-29 cells were evaluated using MTT and scratch wound healing assay, respectively. Quantitative real-time PCR (qRT-PCR) was performed to determine the mRNA expression levels of migration and apoptosis-related genes. Apoptosis and cell cycle distribution were evaluated by flow cytometry. The intracellular level of reactive oxygen species (ROS) was measured using a ROS assay kit. Additionally, matrix metalloproteinase-2/-9 (MMP-2/-9) activity was determined using gelatin zymography. Zerumbone suppressed the viability of the HT-29 cells dose-dependently while having less cytotoxicity on normal NIH/3T3 cells. Zerumbone induced apoptosis in HT-29 cells and arrested the cell cycle in the G2/M phase. These effects were associated with alteration in the expression of apoptosis-related genes (up-regulation of Bax and down-regulation of Bcl-2 genes). Zerumbone also enhanced the generation of ROS in HT-29 cells. Furthermore, zerumbone significantly inhibited the migration of HT-29 cells and decreased MMP-2/-9 mRNA expression and activity. Our findings provide a potential use for zerumbone to induce apoptosis and suppress metastasis in HT-29 cells; thus, it could be developed as a promising natural agent for future CRC therapy.
Collapse
Affiliation(s)
- Fezzeh Memari
- Department of Clinical Biochemistry, Faculty of Medicine, 113380Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, 196469Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Jalili-Nik
- Department of Clinical Biochemistry, Faculty of Medicine, 113380Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, 196469North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Ahmad Ghorbani
- Department of Pharmacology, Faculty of Medicine, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmacological Research Center of Medicinal Plants, 113380Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, 113380Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmacological Research Center of Medicinal Plants, 113380Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Integration of synthetic and natural derivatives revives the therapeutic potential of temozolomide against glioma- an in vitro and in vivo perspective. Life Sci 2022; 301:120609. [DOI: 10.1016/j.lfs.2022.120609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/02/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
|
10
|
Antitumor Effect of the Synthesized Chalcone Analogues on HeLa Cell Line. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2022. [DOI: 10.2478/sjecr-2021-0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Chalcones represent a type of flavonoids which are located at vegetative and reproductive organs of plants and they can be metabolic progenitor molecules for several flavonoids and isoflavonoids. Many studies indicated that molecular structure of chalcone accountable for their anti-tumor, anti-inflammatory and anti-oxidant effects. The aim of our research was to investigate anti-tumor effect and mechanism of action of three synthesized chalcone analogues on HeLa cells. The anti-tumor effectiveness of chalcone analogues was compared to effects of the dehydrozingerone and cisplatin that were used as referent substances.
The viability of the treated cells was evaluated using MTT assay. Evaluation of cell death was determined by flow cytometry and cells were stained with Annexin V-FITC/7-AAD. The result of our research indicated that used chalcones have stronger antitumor effect relative to the dehydrozingerone and cisplatin.
The IC50 values of the chalcones ranged between 1.69-6.18 μM, with CH1 being more cytotoxic after 24 h of treatment, while CH3 being more cytotoxic after 48 h of treatment on HeLa cells. All investigated chalcones induced apoptosis in HeLa cells via mitochondrial pathway, which was detected expression Bax and Bcl- 2 proteins.
Our results provided evidence that chalcones induced apoptosis in HeLa cervical carcinoma through the intrinsic apoptotic pathway. These findings provide insights into the molecular mechanism of chalcones-induced cell death.
Collapse
|
11
|
Matsumoto S, Nakata K, Sagara A, Guan W, Ikenaga N, Ohuchida K, Nakamura M. Efficient pre-treatment for pancreatic cancer using chloroquine-loaded nanoparticles targeting pancreatic stellate cells. Oncol Lett 2021; 22:633. [PMID: 34267825 PMCID: PMC8258615 DOI: 10.3892/ol.2021.12894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic stellate cells (PSCs) play a key role in desmoplastic stroma, which is a characteristic of pancreatic ductal adenocarcinoma (PDAC), and they also enhance the malignancy of pancreatic cancer cells. Our previous study reported chloroquine's mitigating effects on PSC activation; however, the drug is known to induce adverse effects in clinical practice. The present study aimed to reduce chloroquine doses and develop a useful pre-treatment that targets PSCs using nanoparticles. Poly lactic-co-glycolic acid (PLGA) nanoparticles were used as carriers and loaded with indocyanine green (Nano-ICG) or chloroquine (Nano-CQ). Tumor accumulation of Nano-ICG was evaluated using an in vivo imaging system. The effects of chloroquine, Nano-CQ and/or chemotherapy drug gemcitabine were investigated in an orthotopic xenograft mouse model. Nano-ICG selectively accumulated in pancreatic tumors and persisted therein for over 7 days after administration. Additionally, Nano-ICG accumulated in the peritoneal metastasized regions, but not in the liver, kidney and normal pancreatic tissues. Nano-CQ reduced the density of activated PSCs at lower chloroquine doses and significantly restrained tumor progression in combination with gemcitabine. In conclusion, the PLGA nanosystem successfully delivered the drug to pancreatic tumors. Nano-CQ efficiently reduced PSC activation and may be a promising novel pre-treatment strategy for PDAC.
Collapse
Affiliation(s)
- Sokichi Matsumoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akiko Sagara
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Weiyu Guan
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
12
|
Precilla DS, Kuduvalli SS, Purushothaman M, Marimuthu P, Ramachandran MA, Anitha TS. Wnt/β-catenin Antagonists: Exploring New Avenues to Trigger Old Drugs in Alleviating Glioblastoma Multiforme. Curr Mol Pharmacol 2021; 15:338-360. [PMID: 33881978 DOI: 10.2174/1874467214666210420115431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/24/2020] [Accepted: 01/30/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glioblastoma multiforme is one of the most heterogenous primary brain tumor with high mortality. Nevertheless, of the current therapeutic approaches, survival rate remains poor with 12 to 15 months following preliminary diagnosis, this warrants the need for effective treatment modality. Wnt/β-catenin pathway is presumably the most noteworthy pathway up-regulated in almost 80% GBM cases contributing to tumor-initiation, progression and survival. Therefore, therapeutic strategies targeting key components of Wnt/β-catenin cascade using established genotoxic agents like temozolomide and pharmacological inhibitors would be an effective approach to modulate Wnt/β-catenin pathway. Recently, drug repurposing by means of effective combination therapy has gained importance in various solid tumors including GBM, by targeting two or more proteins in a single pathway, thereby possessing the ability to overcome the hurdle implicated by chemo-resistance in GBM. OBJECTIVE In this context, by employing computational tools, an attempt has been carried out to speculate the novel combinations against Wnt/β-catenin signaling pathway. METHODS We have explored the binding interactions of three conventional drugs namely temozolomide, metformin, chloroquine along with three natural compounds viz., epigallocatechin gallate, naringenin and phloroglucinol on the major receptors of Wnt/β-catenin signaling. RESULTS It was noted that all the experimental compounds possessed profound interaction with the two major receptors of Wnt/β-catenin pathway. CONCLUSION To the best of our knowledge, this study is the first of its kind to characterize the combined interactions of the afore-mentioned drugs on Wnt/β-catenin signaling in silico and this will putatively open up new avenues for combination therapies in GBM treatment.
Collapse
Affiliation(s)
- Daisy S Precilla
- Central Inter-Disciplinary Research Facility, School of Biological Sciences, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | - Shreyas S Kuduvalli
- Central Inter-Disciplinary Research Facility, School of Biological Sciences, Sri Balaji Vidyapeeth (Deemed to-be University), Puducherry, India
| | | | - Parthiban Marimuthu
- Structural Bioinformatics Laboratory - Pharmacy, Faculty of Science and Engineering, Åbo Akademi University, Turku. Finland
| | | | | |
Collapse
|
13
|
Li H, Lv J, Guo J, Wang S, Liu S, Ma Y, Liang Z, Wang Y, Qi W, Qiu W. 5-Fluorouracil enhances the chemosensitivity of gastric cancer to TRAIL via inhibition of the MAPK pathway. Biochem Biophys Res Commun 2021; 540:108-115. [PMID: 33476960 DOI: 10.1016/j.bbrc.2021.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 01/04/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has the ability to selectively trigger cancer cell apoptosis and can be used as a target for tumor therapy. However, gastric cancer cells are usually insensitive to TRAIL so reducing this drug resistance may improve the treatment of gastric cancer. In this study, we used Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) experiments to determine the effects of 5-fluorouracil (5-FU) and TRAIL on the proliferation of gastric cancer cells. An Annexin V/propidium iodide (PI) staining experiment was used to detect apoptosis, and Western blotting was used to analyze the expression levels of apoptosis-related proteins and mitogen-activated protein kinase (MAPK) pathway proteins. The antitumor effects of 5-FU and TRAIL were verified in vivo using a nude mouse tumorigenesis experiment, and a TUNEL assay was performed to evaluate apoptosis in tumor tissue from the nude mice. We found the combination of 5-FU and TRAIL had a greater inhibitory effect on the proliferation of gastric cancer cells than 5-FU or TRAIL alone both in vivo and in vitro. 5-FU enhanced TRAIL-induced gastric cancer cell apoptosis by inactivating the MAPK pathway. Overall, our analysis firstly provided new insights into the role of 5-FU in increasing sensitivity to TRAIL. 5-FU can be used as a sensitizer for TRAIL, and its administration is a potential strategy for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Hui Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Jing Lv
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Jing Guo
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Shihai Liu
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Yingji Ma
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Zhiwei Liang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Yunyun Wang
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China
| | - Weiwei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China.
| | - Wensheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong, China.
| |
Collapse
|
14
|
Calabrese EJ, Hanekamp JC, Hanekamp YN, Kapoor R, Dhawan G, Agathokleous E. Chloroquine commonly induces hormetic dose responses. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 755:142436. [PMID: 33017762 PMCID: PMC7518853 DOI: 10.1016/j.scitotenv.2020.142436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 05/02/2023]
Abstract
The use of chloroquine in the treatment of COVID-19 has received considerable attention. The recent intense focus on this application of chloroquine stimulated an investigation into the effects of chloroquine at low doses on highly biologically-diverse models and whether it may induce hormetic-biphasic dose response effects. The assessment revealed that hormetic effects have been commonly induced by chloroquine, affecting numerous cell types, including tumor cell lines (e.g. human breast and colon) and non-tumor cell lines, enhancing viral replication, sperm motility, various behavioral endpoints as well as decreasing risks of convulsions, and enhancing a spectrum of neuroprotective responses within a preconditioning experimental framework. These diverse and complex findings indicate that hormetic dose responses commonly occur with chloroquine treatment with a range of biological models and endpoints. These findings have implications concerning study design features including the number and spacing of doses, and suggest a range of possible clinical concerns and opportunities depending on the endpoint considered.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Sciences, University of Massachusetts, Amherst, MA 01003, United States of America.
| | - Jaap C Hanekamp
- University College Roosevelt, Lange Noordstraat 1, NL-4331 CB Middelburg, the Netherlands.
| | - Yannic N Hanekamp
- University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Rachna Kapoor
- Saint Francis Hospital and Medical Center, Hartford, CT, United States of America
| | - Gaurav Dhawan
- University of Massachusetts, Human Research Protection Office, Research Compliance, University of Massachusetts, Mass Venture Center, Hadley, MA 01035, United States of America
| | - Evgenios Agathokleous
- Key Laboratory of Agrometeorology of Jiangsu Province, Institute of Ecology, School of Applied Meteorology, Nanjing University of Information Science & Technology, Nanjing 210044, China.
| |
Collapse
|
15
|
Lee H, Han JH, Kim S, Kim S, Cho DH, Woo CH. Anti-malarial Drugs Reduce Vascular Smooth Muscle Cell Proliferation via Activation of AMPK and Inhibition of Smad3 Signaling. J Lipid Atheroscler 2020; 8:267-276. [PMID: 32821717 PMCID: PMC7379117 DOI: 10.12997/jla.2019.8.2.267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/30/2019] [Accepted: 09/16/2019] [Indexed: 11/18/2022] Open
Abstract
Objective The aim of this study was to investigate the effects of 2 anti-malarial drugs, chloroquine (CQ) and hydroxychloroquine (HCQ), on inhibition of vascular smooth muscle cell (VSMC) proliferation both in vivo and in vitro via Adenosine monophosphate-activated protein kinase (AMPK) activation. Methods Protein and mRNA levels were determined by western blot analysis and real-time reverse transcription-polymerase chain reaction in primary rat VSMCs treated with CQ and HCQ, respectively. Cell proliferation was measured by flow cytometry and cell counting. Mice carotid arteries were ligated and treated with CQ or HCQ every other day for 3 weeks. Pathological changes of carotid arteries were visualized by both microscopy and fluorescence microscopy. Results CQ and HCQ increase AMPK phosphorylation in VSMCs. Both CQ and HCQ decrease platelet-derived growth factor-induced VSMC proliferation and cell cycle progression in an AMPK-dependent manner. In addition, CQ and HCQ inhibit Smad3 phosphorylation and VSMC proliferation induced by transforming growth factor-β1. Moreover, CQ and HCQ diminished neointimal proliferation in a mouse model of carotid artery ligation-induced neointima formation. Conclusion The results demonstrated that CQ and HCQ inhibit cell proliferation and cell cycle progression in VSMCs via the AMPK-dependent signaling pathway. Carotid artery ligation-induced intima thickness was reduced in mouse arteries treated with CQ or HCQ, suggesting a role for antimalarial drugs in treating atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Heejung Lee
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Jung-Hwa Han
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Sujin Kim
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Suji Kim
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Du-Hyong Cho
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Chang-Hoon Woo
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
16
|
Eloranta K, Cairo S, Liljeström E, Soini T, Kyrönlahti A, Judde JG, Wilson DB, Heikinheimo M, Pihlajoki M. Chloroquine Triggers Cell Death and Inhibits PARPs in Cell Models of Aggressive Hepatoblastoma. Front Oncol 2020; 10:1138. [PMID: 32766148 PMCID: PMC7379510 DOI: 10.3389/fonc.2020.01138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/05/2020] [Indexed: 12/28/2022] Open
Abstract
Background: Hepatoblastoma (HB) is the most common pediatric liver malignancy. Despite advances in chemotherapeutic regimens and surgical techniques, the survival of patients with advanced HB remains poor, underscoring the need for new therapeutic approaches. Chloroquine (CQ), a drug used to treat malaria and rheumatologic diseases, has been shown to inhibit the growth and survival of various cancer types. We examined the antineoplastic activity of CQ in cell models of aggressive HB. Methods: Seven human HB cell models, all derived from chemoresistant tumors, were cultured as spheroids in the presence of relevant concentrations of CQ. Morphology, viability, and induction of apoptosis were assessed after 48 and 96 h of CQ treatment. Metabolomic analysis and RT-qPCR based Death Pathway Finder array were used to elucidate the molecular mechanisms underlying the CQ effect in a 2-dimensional cell culture format. Quantitative western blotting was performed to validate findings at the protein level. Results: CQ had a significant dose and time dependent effect on HB cell viability both in spheroids and in 2-dimensional cell cultures. Following CQ treatment HB spheroids exhibited increased caspase 3/7 activity indicating the induction of apoptotic cell death. Metabolomic profiling demonstrated significant decreases in the concentrations of NAD+ and aspartate in CQ treated cells. In further investigations, oxidation of NAD+ decreased as consequence of CQ treatment and NAD+/NADH balance shifted toward NADH. Aspartate supplementation rescued cells from CQ induced cell death. Additionally, downregulated expression of PARP1 and PARP2 was observed. Conclusions: CQ treatment inhibits cell survival in cell models of aggressive HB, presumably by perturbing NAD+ levels, impairing aspartate bioavailability, and inhibiting PARP expression. CQ thus holds potential as a new agent in the management of HB.
Collapse
Affiliation(s)
- Katja Eloranta
- Pediatric Research Center, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | | | - Emmi Liljeström
- Pediatric Research Center, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Tea Soini
- Pediatric Research Center, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland.,Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Antti Kyrönlahti
- Pediatric Research Center, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | | | - David B Wilson
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO, United States.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Markku Heikinheimo
- Pediatric Research Center, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland.,Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO, United States
| | - Marjut Pihlajoki
- Pediatric Research Center, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| |
Collapse
|
17
|
Zerumbone Promotes Cytotoxicity in Human Malignant Glioblastoma Cells through Reactive Oxygen Species (ROS) Generation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3237983. [PMID: 32454937 PMCID: PMC7225859 DOI: 10.1155/2020/3237983] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/13/2020] [Accepted: 04/23/2020] [Indexed: 01/28/2023]
Abstract
Glioblastoma multiforme (GBM) is the most hostile tumor in the central nervous system. Unfortunately, the prognosis of GBM patients is poor following surgical interventions, chemotherapy, and radiotherapy. Consequently, more efficient and effective treatment options for the treatment of GBM need to be explored. Zerumbone, as a sesquiterpene derived from Zingiber zerumbet Smith, has substantial cytotoxic and antiproliferative activities in some types of cancer. Here, we show that exposure of GBM cells (U-87 MG) to Zerumbone demonstrated significant growth inhibition in a concentration-dependent manner. Zerumbone also induced apoptosis and caused cell cycle arrest of human GBM U-87 MG cells in the G2/M phase of the cell cycle. In detail, the apoptotic process triggered by Zerumbone involved the upregulation of proapoptotic Bax and the suppression of antiapoptotic Bcl-2 genes expression as determined by qRT-PCR. Moreover, Zerumbone enhanced the generation of reactive oxygen species (ROS), and N-acetyl cysteine (NAC), as an antioxidant, reversed the ROS-induced cytotoxicity of U-87 MG cells. The Western blot analysis suggested that Zerumbone activated the NF-κB p65, which was partly inhibited by NAC treatment. Collectively, our results confirmed that Zerumbone induces cytotoxicity by ROS generation. Thus, the study raises the possibility of Zerumbone as a potential natural agent for treating GBM due to its ability to induce cytotoxicity.
Collapse
|
18
|
Lyu X, Zeng L, Zhang H, Ke Y, Liu X, Zhao N, Yuan J, Chen G, Yang S. Hydroxychloroquine suppresses lung tumorigenesis via inducing FoxO3a nuclear translocation through STAT3 inactivation. Life Sci 2020; 246:117366. [PMID: 32001266 DOI: 10.1016/j.lfs.2020.117366] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/09/2020] [Accepted: 01/26/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND Hydroxychloroquine exhibits synergistic anticancer properties as an adjuvant. However, the role and molecular mechanisms underlying of HCQ as monotherapy for lung adenocarcinoma (LUAD) have yet to be elucidated. METHODS We assessed the antitumor effects of HCQ in LUAD cells through a series of in vitro and in vivo assays. GEO database and R packages were used to predict molecular mechanisms of HCQ in the treatment of lung adenocarcinoma, followed by verification of gene expression and subcellular localization via immunoblotting, immunofluorescent and immunohistochemistry assays. RESULTS We showed the phenotypic effects that HCQ inhibited cell growth, induced apoptosis and cell cycle arrest at G1/S transition in A549 and PC-9 cells, which was associated with inhibition of CDK2, CDK4, CyclinD1 and CyclinE, but up-regulation of p21 and p27Kip1. Bioinformatic analysis predicted that 63 targets related to HCQ and LUAD were mainly enriched in JAK-STAT and FoxO pathways. Then, we observed that HCQ decreased the phosphorylation of STAT3, but increased the expression of FoxO3a and its accumulation in the nucleus. The specific STAT3 inhibitor cryptotanshinon augmented the HCQ-induced upregulation and nuclear translocation of FoxO3a. In addition, HCQ increased the expression of p27Kip1, which was impaired by FoxO3a blockade with siRNA. Finally, ablation of p27Kip1 expression abrogated the cytotoxicity of HCQ. More importantly, similar results were further confirmed in vivo. CONCLUSIONS Taken together, this study suggests that STAT3/FoxO3a/p27Kip1 signaling pathway is involved in the anticancer effects of HCQ, and provides preliminary evidence for therapeutic prospects of HCQ alone in LUAD.
Collapse
Affiliation(s)
- Xin Lyu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi, PR China
| | - Lizhong Zeng
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi, PR China
| | - Hua Zhang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi, PR China
| | - Yue Ke
- Department of Radiation Oncology, Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi, PR China
| | - Xuan Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi, PR China
| | - Nannan Zhao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi, PR China
| | - Jingyan Yuan
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi, PR China
| | - Guoan Chen
- School of Medicine, Southern University of Science and Technology, No. 1088, Xueyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi, PR China.
| |
Collapse
|
19
|
Abstract
Malignant melanoma is the most aggressive and notorious skin cancer, and metastatic disease is associated with very poor long-term survival outcomes. Although metastatic melanoma patients with oncogenic mutations in the BRAF gene initially respond well to the treatment with specific BRAF inhibitors, most of them will eventually develop resistance to this targeted therapy. As a highly conserved catabolic process, autophagy is responsible for the maintenance of cellular homeostasis and cell survival, and is involved in multiple diseases, including cancer. Recent study results have indicated that autophagy might play a decisive role in the resistance to BRAF inhibitors in BRAF-mutated melanomas. In this review, we will discuss how autophagy is up-regulated by BRAF inhibitors, and how autophagy induces the resistance to these agents.
Collapse
|
20
|
Natural Product Mediated Regulation of Death Receptors and Intracellular Machinery: Fresh from the Pipeline about TRAIL-Mediated Signaling and Natural TRAIL Sensitizers. Int J Mol Sci 2019; 20:ijms20082010. [PMID: 31022877 PMCID: PMC6515249 DOI: 10.3390/ijms20082010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/24/2022] Open
Abstract
Rapidly developing resistance against different therapeutics is a major stumbling block in the standardization of therapy. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-mediated signaling has emerged as one of the most highly and extensively studied signal transduction cascade that induces apoptosis in cancer cells. Rapidly emerging cutting-edge research has helped us to develop a better understanding of the signaling machinery involved in inducing apoptotic cell death. However, excitingly, cancer cells develop resistance against TRAIL-induced apoptosis through different modes. Loss of cell surface expression of TRAIL receptors and imbalance of stoichiometric ratios of pro- and anti-apoptotic proteins play instrumental roles in rewiring the machinery of cancer cells to develop resistance against TRAIL-based therapeutics. Natural products have shown excellent potential to restore apoptosis in TRAIL-resistant cancer cell lines and in mice xenografted with TRAIL-resistant cancer cells. Significantly refined information has previously been added and continues to enrich the existing pool of knowledge related to the natural-product-mediated upregulation of death receptors, rebalancing of pro- and anti-apoptotic proteins in different cancers. In this mini review, we will set spotlight on the most recently published high-impact research related to underlying mechanisms of TRAIL resistance and how these deregulations can be targeted by natural products to restore TRAIL-mediated apoptosis in different cancers.
Collapse
|
21
|
Qin A, Zhang Q, Wang J, Sayeed I, Stein DG. Is a combination of progesterone and chloroquine more effective than either alone in the treatment of cerebral ischemic injury? Restor Neurol Neurosci 2019; 37:1-10. [PMID: 30741704 DOI: 10.3233/rnn-180837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND In this proof-of-concept paper, we investigated whether combination treatment with progesterone (P4) and chloroquine (CQ) would reduce ischemic injury more effectively than either agent alone in a transient middle cerebral artery occlusion (tMCAO) model in male rats. METHODS P4 (8 mg/kg) and CQ (25 mg/kg) were given alone or in combination beginning at different times during surgery and for 3 days post-occlusion. Locomotor activity and grip strength were evaluated as measures of impairment and recovery. Infarct size was assessed by TTC staining. Markers of autophagy (LC3 and SQSTM1/p62) and apoptosis (Bcl-2 and Bax) were evaluated with western blotting. RESULTS At the doses we employed, the combination was not more effective than either drug given separately on measures of grip strength or locomotor activity. However, combination therapy substantially reduced infarct size, and significantly increased Bcl-2 protein levels and suppressed Bax expression. Progesterone decreased the expression of LC3-II 24 h and SQSTM1/p62 after ischemia. CONCLUSIONS Our findings suggest that combination therapy with P4 and CQ is not detrimental and has a small-to-moderate additive neuroprotective effect on ischemic injury in rats without substantively affecting behavioral outcomes. CQ and P4 may help to regulate the expression of both autophagy-related and apoptosis-related proteins.
Collapse
Affiliation(s)
- Aiping Qin
- Department of Pharmacy, Jiangsu Health Vocational College, Nanjing, Jiangsu, China
| | - Qian Zhang
- Xuzhou Medical University, Xuzhou, China
| | - Jun Wang
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, Georgia, USA
| | - Iqbal Sayeed
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, Georgia, USA
| | - Donald G Stein
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
22
|
Twomey JD, Zhang B. Circulating Tumor Cells Develop Resistance to TRAIL-Induced Apoptosis Through Autophagic Removal of Death Receptor 5: Evidence from an In Vitro Model. Cancers (Basel) 2019; 11:cancers11010094. [PMID: 30650534 PMCID: PMC6356356 DOI: 10.3390/cancers11010094] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 01/03/2023] Open
Abstract
Circulating tumor cells (CTCs) in the peripheral blood are the precursors to distant metastasis but the underlying mechanisms are poorly understood. This study aims at understanding the molecular features within CTCs, in relation to their metastatic potential. Using in vitro CTC models, in which breast cancer cell lines were cultured in non-adherent conditions simulating the microenvironment in the blood stream, we found that the suspension culture resulted in resistance to TNF-related apoptosis inducing ligand (TRAIL)-mediated cell death. Such a resistance was directly correlated with a reduction in surface and total levels of DR5 protein. In the non-adherent state, the cells underwent a rapid autophagic flux, characterized by an accumulation of autophagosome organelles. Notably, DR5 was translocated to the autophagosomes and underwent a lysosomal degradation. Our data suggest that CTCs may evade the TNF cytokine-mediated immune surveillance through a downregulation of the death receptor (DR) expression. The data warrants further studies in cancer patients to find the status of DRs and other molecular features within primary CTCs, in relation to disease progression or chemoresistance.
Collapse
Affiliation(s)
- Julianne D Twomey
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| |
Collapse
|
23
|
Papanagnou P, Papadopoulos GE, Stivarou T, Pappas A. Toward fully exploiting the therapeutic potential of marketed pharmaceuticals: the use of octreotide and chloroquine in oncology. Onco Targets Ther 2018; 12:319-339. [PMID: 30643430 PMCID: PMC6317484 DOI: 10.2147/ott.s182685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pleiotropy in biological systems and their targeting allows many pharmaceuticals to be used for multiple therapeutic purposes. Fully exploiting the therapeutic properties of drugs that are already marketed would be highly advantageous. This is especially the case in the field of oncology, where the ineffectiveness of typical anticancer agents is a common issue, while the development of novel anticancer agents is a costly and particularly time-consuming process. Octreotide and chloroquine are two pharmaceuticals that exhibit profound antitumorigenic activities. However, the current therapeutic use of octreotide is restricted primarily to the management of acromegaly and neuroendocrine tumors, both of which are rare medical conditions. Similarly, chloroquine is used mainly for the treatment of malaria, which is designated as a rare disease in Western countries. This limited exploitation contradicts the experimental findings of numerous studies outlining the possible expansion of the use of octreotide to include the treatment of common human malignancies and the repositioning of chloroquine in oncology. Herein, we review the current knowledge on the antitumor function of these two agents stemming from preclinical or clinical experimentation. In addition, we present in silico evidence on octreotide potentially binding to multiple Wnt-pathway components. This will hopefully aid in the design of new efficacious anticancer therapeutic regimens with minimal toxicity, which represents an enormous unmet demand in oncology.
Collapse
Affiliation(s)
| | | | - Theodora Stivarou
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, Athens, Greece
| | - Anastasios Pappas
- Department of Urology, Agios Savvas Cancer Hospital, Athens 11522, Greece,
| |
Collapse
|
24
|
Chloroquine Urea Derivatives: Synthesis and Antitumor Activity in Vitro. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2018; 68:471-483. [PMID: 31259711 DOI: 10.2478/acph-2018-0039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/14/2018] [Indexed: 12/30/2022]
Abstract
In the current paper, we describe the design, synthesis and antiproliferative screening of novel chloroquine derivatives with a quinoline core linked to a hydroxy or halogen amine through a flexible aminobutyl chain and urea spacer. Synthetic pathway leading to chloroquine urea derivatives 4-10 includes two crucial steps: i) synthesis of chloroquine benzotriazolide 3 and ii) formation of urea derivatives through the reaction of compound 3 with the corresponding amine. Testing of antiproliferative activity against four human cancer cell lines revealed that chloroquine urea derivatives 9 and 10 with aromatic moieties show activity at micromolar concentrations. Therefore, these molecules represent interesting lead compounds that might provide an insight into the design of new anticancer agents.
Collapse
|
25
|
Dower CM, Bhat N, Gebru MT, Chen L, Wills CA, Miller BA, Wang HG. Targeted Inhibition of ULK1 Promotes Apoptosis and Suppresses Tumor Growth and Metastasis in Neuroblastoma. Mol Cancer Ther 2018; 17:2365-2376. [PMID: 30166400 DOI: 10.1158/1535-7163.mct-18-0176] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/20/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022]
Abstract
Neuroblastoma is the most common extracranial solid malignancy in the pediatric population, accounting for over 9% of all cancer-related deaths in children. Autophagy is a cell self-protective mechanism that promotes tumor cell growth and survival, making it an attractive target for treating cancer. However, the role of autophagy in neuroblastoma tumor growth and metastasis is largely undefined. Here we demonstrate that targeted inhibition of an essential autophagy kinase, unc-51 like autophagy kinase 1 (ULK1), with a recently developed small-molecule inhibitor of ULK1, SBI-0206965, significantly reduces cell growth and promotes apoptosis in SK-N-AS, SH-SY5Y, and SK-N-DZ neuroblastoma cell lines. Furthermore, inhibition of ULK1 by a dominant-negative mutant of ULK1 (dnULK1K46N) significantly reduces growth and metastatic disease and prolongs survival of mice bearing SK-N-AS xenograft tumors. We also show that SBI-0206965 sensitizes SK-N-AS cells to TRAIL treatment, but not to mTOR inhibitors (INK128, Torin1) or topoisomerase inhibitors (doxorubicin, topotecan). Collectively, these findings demonstrate that ULK1 is a viable drug target and suggest that inhibitors of ULK1 may provide a novel therapeutic option for the treatment of neuroblastoma. Mol Cancer Ther; 17(11); 2365-76. ©2018 AACR.
Collapse
Affiliation(s)
- Christopher M Dower
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Neema Bhat
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Melat T Gebru
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Longgui Chen
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Carson A Wills
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Barbara A Miller
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Hong-Gang Wang
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
| |
Collapse
|