1
|
Li J, Pan J, Wang L, Ji G, Dang Y. Colorectal Cancer: Pathogenesis and Targeted Therapy. MedComm (Beijing) 2025; 6:e70127. [PMID: 40060193 PMCID: PMC11885891 DOI: 10.1002/mco2.70127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 04/29/2025] Open
Abstract
Colorectal cancer (CRC) ranks among the most prevalent malignant neoplasms globally. A growing body of evidence underscores the pivotal roles of genetic alterations and dysregulated epigenetic modifications in the pathogenesis of CRC. In recent years, the reprogramming of tumor cell metabolism has been increasingly acknowledged as a hallmark of cancer. Substantial evidence suggests a crosstalk between tumor cell metabolic reprogramming and epigenetic modifications, highlighting a complex interplay between metabolism and the epigenetic genome that warrants further investigation. Biomarkers associated with the pathogenesis and metabolic characteristics of CRC hold significant clinical implications. Nevertheless, elucidating the genetic, epigenetic, and metabolic landscapes of CRC continues to pose considerable challenges. Here, we attempt to summarize the key genes driving the onset and progression of CRC and the related epigenetic regulators, clarify the roles of gene expression and signaling pathways in tumor metabolism regulation, and explore the potential crosstalk between epigenetic events and tumor metabolic reprogramming, providing a comprehensive mechanistic explanation for the malignant progression of CRC. Finally, by integrating reliable targets from genetics, epigenetics, and metabolic processes that hold promise for translation into clinical practice, we aim to offer more strategies to overcome the bottlenecks in CRC treatment.
Collapse
Affiliation(s)
- Jingyuan Li
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiashu Pan
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Lisheng Wang
- Department of BiochemistryMicrobiology and ImmunologyFaculty of MedicineUniversity of OttawaOttawaOntarioCanada
- China‐Canada Centre of Research for Digestive DiseasesUniversity of OttawaOttawaOntarioCanada
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
2
|
Kuppusamy P, Haque MM, Traub RJ, Melemedjian OK. Targeting metabolic pathways alleviates bortezomib-induced neuropathic pain without compromising anticancer efficacy in a sex-specific manner. FRONTIERS IN PAIN RESEARCH 2024; 5:1424348. [PMID: 38979441 PMCID: PMC11228363 DOI: 10.3389/fpain.2024.1424348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/10/2024] [Indexed: 07/10/2024] Open
Abstract
Introduction Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating side effect of cancer treatment that significantly impacts patients' quality of life. This study investigated the effects of targeting metabolic pathways on bortezomib-induced neuropathic pain and tumor growth using a Lewis lung carcinoma (LLC) mouse model, while exploring potential sex differences. Methods Male and female C57BL/6J mice were implanted with LLC cells and treated with bortezomib alone or in combination with metformin, dichloroacetate (DCA), or oxamate. Tactile allodynia was assessed using von Frey filaments. Tumor volume and weight were measured to evaluate tumor growth. Results Metformin, DCA, and oxamate effectively attenuated bortezomib-induced neuropathic pain without compromising the anticancer efficacy of bortezomib in both male and female mice. The LLC model exhibited a paraneoplastic neuropathy-like phenotype. Significant sex differences were observed, with male mice exhibiting larger tumors compared to females. Oxamate was more effective in alleviating allodynia in males, while metformin and DCA showed greater efficacy in reducing tumor growth in females. Discussion Targeting metabolic pathways can alleviate CIPN without interfering with bortezomib's anticancer effects. The LLC model may serve as a tool for studying paraneoplastic neuropathy. Sex differences in tumor growth and response to metabolic interventions highlight the importance of considering sex as a biological variable in preclinical and clinical studies investigating cancer biology, CIPN, and potential therapeutic interventions.
Collapse
Affiliation(s)
- Panjamurthy Kuppusamy
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Md Mamunul Haque
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Richard J. Traub
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
- UM Center to Advance Chronic Pain Research, Baltimore, MD, United States
| | - Ohannes K. Melemedjian
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
- UM Center to Advance Chronic Pain Research, Baltimore, MD, United States
- UM Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
3
|
Koltai T, Fliegel L. Dichloroacetate for Cancer Treatment: Some Facts and Many Doubts. Pharmaceuticals (Basel) 2024; 17:744. [PMID: 38931411 PMCID: PMC11206832 DOI: 10.3390/ph17060744] [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: 03/28/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Rarely has a chemical elicited as much controversy as dichloroacetate (DCA). DCA was initially considered a dangerous toxic industrial waste product, then a potential treatment for lactic acidosis. However, the main controversies started in 2008 when DCA was found to have anti-cancer effects on experimental animals. These publications showed contradictory results in vivo and in vitro such that a thorough consideration of this compound's in cancer is merited. Despite 50 years of experimentation, DCA's future in therapeutics is uncertain. Without adequate clinical trials and health authorities' approval, DCA has been introduced in off-label cancer treatments in alternative medicine clinics in Canada, Germany, and other European countries. The lack of well-planned clinical trials and its use by people without medical training has discouraged consideration by the scientific community. There are few thorough clinical studies of DCA, and many publications are individual case reports. Case reports of DCA's benefits against cancer have been increasing recently. Furthermore, it has been shown that DCA synergizes with conventional treatments and other repurposable drugs. Beyond the classic DCA target, pyruvate dehydrogenase kinase, new target molecules have also been recently discovered. These findings have renewed interest in DCA. This paper explores whether existing evidence justifies further research on DCA for cancer treatment and it explores the role DCA may play in it.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires 2199, Argentina
| | - Larry Fliegel
- Department of Biochemistry, University Alberta, Edmonton, AB T6G 2H7, Canada;
| |
Collapse
|
4
|
Jo MY, Jeong YJ, Song KH, Choi YH, Kwon TK, Chang YC. 4-O-Methylascochlorin Synergistically Enhances 5-Fluorouracil-Induced Apoptosis by Inhibiting the Wnt/β-Catenin Signaling Pathway in Colorectal Cancer Cells. Int J Mol Sci 2024; 25:5746. [PMID: 38891932 PMCID: PMC11172374 DOI: 10.3390/ijms25115746] [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: 04/22/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
4-O-Methyl-ascochlorin (MAC), a derivative of the prenyl-phenol antibiotic ascochlorin extracted from the fungus Ascochyta viciae, shows anticarcinogenic effects on various cancer cells. 5-Fluorouracil (5-FU) is used to treat colorectal cancer (CRC); however, its efficacy must be enhanced. In this study, we investigated the molecular mechanisms by which MAC acts synergistically with 5-FU to inhibit cell proliferation and induce apoptosis in CRC cells. MAC enhanced the cytotoxic effects of 5-FU by suppressing the Akt/mTOR/p70S6K and Wnt/β-catenin signaling pathways. It also reduced the viability of 5-FU-resistant (5-FU-R) cells. Furthermore, expression of anti-apoptosis-related proteins and cancer stem-like cell (CSC) markers by 5-FU-R cells decreased in response to MAC. Similar to MAC, the knockdown of CTNNB1 induced apoptosis and reduced expression of mRNA encoding CRC markers in 5-FU-R cells. In summary, these results suggest that MAC and other β-catenin modulators may be useful in overcoming the 5-FU resistance of CRC cells.
Collapse
Affiliation(s)
- Min-Young Jo
- Research Institute of Biomedical Engineering and Department of Cell Biology, Daegu Catholic University School of Medicine, Daegu 42472, Republic of Korea
| | - Yun-Jeong Jeong
- Research Institute of Biomedical Engineering and Department of Cell Biology, Daegu Catholic University School of Medicine, Daegu 42472, Republic of Korea
| | - Kwon-Ho Song
- Research Institute of Biomedical Engineering and Department of Cell Biology, Daegu Catholic University School of Medicine, Daegu 42472, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Cell Biology, Daegu Catholic University School of Medicine, Daegu 42472, Republic of Korea
| |
Collapse
|
5
|
Li Y, Xie Z, Lei X, Yang X, Huang S, Yuan W, Deng X, Wang Z, Tang G. Recent advances in pyruvate dehydrogenase kinase inhibitors: Structures, inhibitory mechanisms and biological activities. Bioorg Chem 2024; 144:107160. [PMID: 38301426 DOI: 10.1016/j.bioorg.2024.107160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/03/2024]
Abstract
Metabolism is reprogrammed in a variety of cancer cells to ensure their rapid proliferation. Cancer cells prefer to utilize glycolysis to produce energy as well as to provide large amounts of precursors for their division. In this process, cancer cells inhibit the activity of pyruvate dehydrogenase complex (PDC) by upregulating the expression of pyruvate dehydrogenase kinases (PDKs). Inhibiting the activity of PDKs in cancer cells can effectively block this metabolic transition in cancer cells, while also activating mitochondrial oxidative metabolism and promoting apoptosis of cancer cells. To this day, the study of PDKs inhibitors has become one of the research hotspots in the field of medicinal chemistry. Novel structures targeting PDKs are constantly being discovered, and some inhibitors have entered the clinical research stage. Here, we reviewed the research progress of PDKs inhibitors in recent years and classified them according to the PDKs binding sites they acted on, aiming to summarize the structural characteristics of inhibitors acting on different binding sites and explore their clinical application value. Finally, the shortcomings of some PDKs inhibitors and the further development direction of PDKs inhibitors are discussed.
Collapse
Affiliation(s)
- Yiyang Li
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhizhong Xie
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyong Lei
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyan Yang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Sheng Huang
- Jiuzhitang Co., Ltd, Changsha, Hunan 410007, China
| | - Weixi Yuan
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiangping Deng
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
6
|
Hu J, Li A, Guo Y, Ma T, Feng S. The relationship between tumor metabolism and 5-fluorouracil resistance. Biochem Pharmacol 2023; 218:115902. [PMID: 37922975 DOI: 10.1016/j.bcp.2023.115902] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Affiliation(s)
- Jingyi Hu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Anqi Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yueyang Guo
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ting Ma
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Siqi Feng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
7
|
Mironiuk-Puchalska E, Karatsai O, Żuchowska A, Wróblewski W, Borys F, Lehka L, Rędowicz MJ, Koszytkowska-Stawińska M. Development of 5-fluorouracil-dichloroacetate mutual prodrugs as anticancer agents. Bioorg Chem 2023; 140:106784. [PMID: 37639758 DOI: 10.1016/j.bioorg.2023.106784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
5-Fluorouracil (5-FU) is one of the most widely applied chemotherapeutic agents with a broad spectrum of activity. However, despite this versatile activity, its use poses many limitations. Herein, novel derivatives of 5-FU and dichloroacetic acid have been designed and synthesized as a new type of codrugs, also known as mutual prodrugs, to overcome the drawbacks of 5-FU and enhance its therapeutic efficiency. The stability of the obtained compounds has been tested at various pH values using different analytical techniques, namely HPLC and potentiometry. The antiproliferative activity of the new 5-FU derivatives was assessed in vitro on SK-MEL-28 and WM793 human melanoma cell lines in 2D culture as well as on A549 human lung carcinoma, MDA-MB-231 breast adenocarcinoma, LL24 normal lung tissue, and HMF normal breast tissue as a multicellular 3D spheroid model cultured in standard (static) conditions and with the use of microfluidic systems, which to a great extent resembles the in vivo environment. In all cases, new mutual prodrugs showed a higher cytotoxic activity toward cancer models and lower to normal cell models than the parent 5-FU itself.
Collapse
Affiliation(s)
- Ewa Mironiuk-Puchalska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland.
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Agnieszka Żuchowska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Wojciech Wróblewski
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Filip Borys
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Lilya Lehka
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | | |
Collapse
|
8
|
Xu L, Wu X, Liu H, Dong G, Zhan J, Li G, Wang G, Liu T. Effects of combination docetaxel with NO treatment to enhance the anti-nasopharyngeal carcinoma efficiency in vitro and in vivo. Eur J Pharm Sci 2022; 178:106281. [PMID: 35995348 DOI: 10.1016/j.ejps.2022.106281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/05/2022] [Accepted: 08/18/2022] [Indexed: 02/05/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is one of the major causes of death in Southern China. Due to the insidious location of NPC, the therapeutic effect of locoregionally advanced NPC is still unsatisfactory. In this work, to improve the treatment efficiency, combining DOC and JS-K to inhibit NPC cells (HNE-1) in vitro was investigated, as well as its possible mechanisms. Moreover, the in vivo effects of DOC and JS-K combination treatment were also evaluated in a xenograft model with HNE-1 cells. In vitro experiments including cell proliferation, migration ability, apoptosis, and expression levels of apoptosis-associated proteins revealed that the combination of DOC and JS-K was able to enhance antitumor effects. In vivo results further confirmed a significant treatment effect without obvious toxicity on mice. The present work provides a promising idea for the treatment of locally advanced NPC.
Collapse
Affiliation(s)
- Lingling Xu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xidong Wu
- Department of Drug Safety Evaluation, Jiangxi Testing Center of Medical Instruments, Nanchang, 330029, China
| | - Huiqin Liu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; Shantou University Medical College, Shantou, 515063, China
| | - Guangyuan Dong
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; Shantou University Medical College, Shantou, 515063, China
| | - Jiandong Zhan
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Guanxue Li
- Pediatric Critical Care Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280, China
| | - Guanhai Wang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Tao Liu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
9
|
Aputen AD, Elias MG, Gilbert J, Sakoff JA, Gordon CP, Scott KF, Aldrich-Wright JR. Bioactive Platinum(IV) Complexes Incorporating Halogenated Phenylacetates. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27207120. [PMID: 36296713 PMCID: PMC9611758 DOI: 10.3390/molecules27207120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
Abstract
A new series of cytotoxic platinum(IV) complexes (1-8) incorporating halogenated phenylacetic acid derivatives (4-chlorophenylacetic acid, 4-fluorophenylacetic acid, 4-bromophenylacetic acid and 4-iodophenylacetic acid) were synthesised and characterised using spectroscopic and spectrometric techniques. Complexes 1-8 were assessed on a panel of cell lines including HT29 colon, U87 glioblastoma, MCF-7 breast, A2780 ovarian, H460 lung, A431 skin, Du145 prostate, BE2-C neuroblastoma, SJ-G2 glioblastoma, MIA pancreas, the ADDP-resistant ovarian variant, and the non-tumour-derived MCF10A breast line. The in vitro cytotoxicity results confirmed the superior biological activity of the studied complexes, especially those containing 4-fluorophenylacetic acid and 4-bromophenylacetic acid ligands, namely 4 and 6, eliciting an average GI50 value of 20 nM over the range of cell lines tested. In the Du145 prostate cell line, 4 exhibited the highest degree of potency amongst the derivatives, displaying a GI50 value of 0.7 nM, which makes it 1700-fold more potent than cisplatin (1200 nM) and nearly 7-fold more potent than our lead complex, 56MESS (4.6 nM) in this cell line. Notably, in the ADDP-resistant ovarian variant cell line, 4 (6 nM) was found to be almost 4700-fold more potent than cisplatin. Reduction reaction experiments were also undertaken, along with studies aimed at determining the complexes' solubility, stability, lipophilicity, and reactive oxygen species production.
Collapse
Affiliation(s)
- Angelico D. Aputen
- School of Science, Western Sydney University, Locked Bag 1797, Sydney, NSW 2751, Australia
| | - Maria George Elias
- School of Science, Western Sydney University, Locked Bag 1797, Sydney, NSW 2751, Australia
- Ingham Institute, Sydney, NSW 2170, Australia
| | - Jayne Gilbert
- Calvary Mater Newcastle Hospital, Newcastle, NSW 2298, Australia
| | | | - Christopher P. Gordon
- School of Science, Western Sydney University, Locked Bag 1797, Sydney, NSW 2751, Australia
| | | | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Locked Bag 1797, Sydney, NSW 2751, Australia
- Correspondence: ; Tel.: +61-246203218
| |
Collapse
|
10
|
Lam SK, Yan S, Lam JSM, Feng Y, Khan M, Chen C, Ko FCF, Ho JCM. Disturbance of the Warburg effect by dichloroacetate and niclosamide suppresses the growth of different sub-types of malignant pleural mesothelioma in vitro and in vivo. Front Pharmacol 2022; 13:1020343. [PMID: 36304150 PMCID: PMC9592830 DOI: 10.3389/fphar.2022.1020343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Inhalation of asbestos fibers is the most common cause of malignant pleural mesothelioma (MPM). In 2004, the United States Food and Drug Administration approved a combination of cisplatin with pemetrexed to treat unresectable MPM. Nonetheless novel treatment is urgently needed. The objective of this study is to report the combination effect of dichloroacetate (DCA) or niclosamide (Nic) Nic in MPM. Materials and methods: The effect of a combination of DCA and Nic was studied using a panel of MPM cell lines (H28, MSTO-211H, H226, H2052, and H2452). Cell viability was monitored by MTT assay. Glycolysis, oxidative phosphorylation, glucose, glycogen, pyruvate, lactate, citrate, succinate and ATP levels were determined by corresponding ELISA. Apoptosis, mitochondrial transmembrane potential, cell cycle analysis, hydrogen peroxide and superoxide were investigated by flow cytometry. Cell migration and colony formation were investigated by transwell migration and colony formation assays respectively. The in vivo effect was confirmed using 211H and H226 nude mice xenograft models. Results and conclusion: Cell viability was reduced. Disturbance of glycolysis and/or oxidative phosphorylation resulted in downregulation of glycogen, citrate and succinate. DCA and/or Nic increased apoptosis, mitochondrial transmembrane depolarization, G2/M arrest and reactive oxygen species. Moreover, DCA and/or Nic suppressed cell migration and colony formation. Furthermore, a better initial tumor suppressive effect was induced by the DCA/Nic combination compared with either drug alone in both 211H and H226 xenograft models. In H226 xenografts, DCA/Nic increased median survival of mice compared with single treatment. Single drug and/or a combination disturbed the Warburg effect and activated apoptosis, and inhibition of migration and proliferation in vivo. In conclusion, dichloroacetate and/or niclosamide showed a tumor suppressive effect in MPM in vitro and in vivo, partially mediated by disturbance of glycolysis/oxidative phosphorylation, apoptosis, ROS production, G2/M arrest, and suppression of migration and proliferation.
Collapse
|
11
|
Hossain M, Roth S, Dimmock JR, Das U. Cytotoxic derivatives of dichloroacetic acid and some metal complexes. Arch Pharm (Weinheim) 2022; 355:e2200236. [DOI: 10.1002/ardp.202200236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/27/2022] [Accepted: 07/30/2022] [Indexed: 11/12/2022]
Affiliation(s)
| | - Shayne Roth
- School of Sciences Indiana University Kokomo Kokomo Indiana USA
| | - Jonathan R. Dimmock
- Drug Discovery and Development Research Cluster University of Saskatchewan Saskatoon Saskatchewan Canada
| | - Umashankar Das
- Drug Discovery and Development Research Cluster University of Saskatchewan Saskatoon Saskatchewan Canada
| |
Collapse
|
12
|
Liu X, Zhao Y, Wu X, Liu Z, Liu X. A novel strategy to fuel cancer immunotherapy: targeting glucose metabolism to remodel the tumor microenvironment. Front Oncol 2022; 12:931104. [PMID: 35924168 PMCID: PMC9340371 DOI: 10.3389/fonc.2022.931104] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/27/2022] [Indexed: 12/20/2022] Open
Abstract
The promising results of immunotherapy in tumors have changed the current treatment modality for cancer. However, the remarkable responses are limited to a minority of patients, which is due to immune suppression in the tumor microenvironment (TME). These include the pre-exists of suppressive immune cells, physical barriers to immune infiltration, antigen and antigen presentation deficiency, and expression of inhibitory immune checkpoint molecules. Recently, increasing evidence reveal that tumor metabolism, especially abnormal glucose metabolism of tumors, plays an essential role in tumor immune escape and is a potential target to combine with immunotherapy. By glucose uptake, tumor cells alter their metabolism to facilitate unregulated cellular proliferation and survival and regulate the expression of inhibitory immune checkpoint molecules. Meanwhile, glucose metabolism also regulates the activation, differentiation, and functions of immunocytes. In addition, tumor mainly utilizes glycolysis for energy generation and cellular proliferation, which cause the TME to deplete nutrients for infiltrating immune cells such as T cells and produce immunosuppressive metabolites. Thus, therapeutics that target glucose metabolism, such as inhibiting glycolytic activity, alleviating hypoxia, and targeting lactate, have shown promise as combination therapies for different types of cancer. In this review, we summarized the functions of glucose metabolism in the tumor cells, immune cells, and tumor microenvironment, as well as strategies to target glucose metabolism in combination with immune checkpoint blockade for tumor therapy.
Collapse
Affiliation(s)
- Xu Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Head, Neck and Mammary Gland Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yujie Zhao
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Wu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihui Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowei Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xiaowei Liu,
| |
Collapse
|
13
|
Al-Azawi A, Sulaiman S, Arafat K, Yasin J, Nemmar A, Attoub S. Impact of Sodium Dichloroacetate Alone and in Combination Therapies on Lung Tumor Growth and Metastasis. Int J Mol Sci 2021; 22:ijms222212553. [PMID: 34830434 PMCID: PMC8624089 DOI: 10.3390/ijms222212553] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 01/07/2023] Open
Abstract
Metabolic reprogramming has been recognized as an essential emerging cancer hallmark. Dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase (PDK), has been reported to have anti-cancer effects by reversing tumor-associated glycolysis. This study was performed to explore the anti-cancer potential of DCA in lung cancer alone and in combination with chemo- and targeted therapies using two non-small cell lung cancer (NSCLC) cell lines, namely, A549 and LNM35. DCA markedly caused a concentration- and time-dependent decrease in the viability and colony growth of A549 and LNM35 cells in vitro. DCA also reduced the growth of tumor xenografts in both a chick embryo chorioallantoic membrane and nude mice models in vivo. Furthermore, DCA decreased the angiogenic capacity of human umbilical vein endothelial cells in vitro. On the other hand, DCA did not inhibit the in vitro cellular migration and invasion and the in vivo incidence and growth of axillary lymph nodes metastases in nude mice. Treatment with DCA did not show any toxicity in chick embryos and nude mice. Finally, we demonstrated that DCA significantly enhanced the anti-cancer effect of cisplatin in LNM35. In addition, the combination of DCA with gefitinib or erlotinib leads to additive effects on the inhibition of LNM35 colony growth after seven days of treatment and to synergistic effects on the inhibition of A549 colony growth after 14 days of treatment. Collectively, this study demonstrates that DCA is a safe and promising therapeutic agent for lung cancer.
Collapse
Affiliation(s)
- Aya Al-Azawi
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates; (A.A.-A.); (S.S.); (K.A.)
| | - Shahrazad Sulaiman
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates; (A.A.-A.); (S.S.); (K.A.)
| | - Kholoud Arafat
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates; (A.A.-A.); (S.S.); (K.A.)
| | - Javed Yasin
- Department of Medicine, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates;
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates;
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates
| | - Samir Attoub
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates; (A.A.-A.); (S.S.); (K.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates
- Institut National de la Santé et de la Recherche Médicale (INSERM), 75013 Paris, France
- Correspondence:
| |
Collapse
|
14
|
Ferns GA, Shahini Shams Abadi M, Raeisi A, Arjmand MH. The Potential Role of Changes in the Glucose and Lipid Metabolic Pathways in Gastrointestinal Cancer Progression: Strategy in Cancer Therapy. Gastrointest Tumors 2021; 8:169-176. [PMID: 34722470 DOI: 10.1159/000517771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/10/2021] [Indexed: 11/19/2022] Open
Abstract
Background Changes in cell metabolism are a well-known feature of some cancers, and this may be involved in the etiology of tumor formation and progression, as well as tumor heterogeneity. These changes may affect fatty acid metabolism and glycolysis and are required to provide the increase in energy necessary for the high rate of proliferation of cancer cells. Gastrointestinal cancers remain a difficult-to-treat cancer, particularly as they are usually diagnosed at a late stage of disease and are associated with poor outcomes. Summary Recently, the changes in the metabolic pathways, including the expression of the rate-limiting enzymes involved, have been considered to be a potential target for therapy for gastrointestinal tumors. Key Message A combination of routine chemotherapy drugs with metabolic inhibitors may improve the effectiveness of treatment.
Collapse
Affiliation(s)
- Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Brighton, United Kingdom
| | - Milad Shahini Shams Abadi
- Department of Microbiology and Immunology, Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ahmad Raeisi
- Clinical Research Development Unit, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad-Hassan Arjmand
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
15
|
Nikravesh H, Khodayar MJ, Behmanesh B, Mahdavinia M, Teimoori A, Alboghobeish S, Zeidooni L. The combined effect of dichloroacetate and 3-bromopyruvate on glucose metabolism in colorectal cancer cell line, HT-29; the mitochondrial pathway apoptosis. BMC Cancer 2021; 21:903. [PMID: 34364387 PMCID: PMC8349486 DOI: 10.1186/s12885-021-08564-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/07/2021] [Indexed: 01/07/2023] Open
Abstract
Background 5-Fluorouracil (5-FU) is regarded as the first line treatment for colorectal cancer; however, its effectiveness is limited by drug resistance. The ultimate goal of cancer therapy is induction of cancer cell death to achieve an effective outcome with minimal side effects. The present work aimed to assess the anti-cancer activities of mitocans which can be considered as an effective anticancer drug due to high specificity in targeting cancer cells. Methods MTT (3–4,5-dimethylthiazol-2-yl-2,5-diphenyltetrazolium bromide) assay was performed to determine the effects of our mitocans on cell viability and cell death. Apoptosis and necrosis, caspase 3 activity, mitochondrial membrane potential and ROS production in HT29 cell lines were analyzed by ApopNexin™ FITC/PI Kit, Caspase- 3 Assay Kit, MitoTracker Green and DCFH-DA, respectively. Moreover, quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expression level of pro-apoptotic (Bax) and anti-apoptotic (Bcl-2) genes in HT29 cell lines. Results Treatment with mitocans (3Br-P + DCA) inhibited the growth of HT29. Moreover, 3Br-P + DCA significantly induced apoptosis and necrosis, activation of caspase 3 activity, depolarize the mitochondrial membrane potential, and ROS production. At a molecular level, 3Br-P + DCA treatment remarkably down-regulated the expression of Bcl-2, while up-regulated the expression of Bax. Conclusion Mitocans, in particular the combined drug, 3Br-P + DCA, could be regarded and more evaluated as a safe and effective compound for CRC treatment. Targeting hexokinase and pyruvate dehydrogenase kinase enzymes may be an option to overcome 5-FU -mediated chemo-resistant in colorectal cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08564-3.
Collapse
Affiliation(s)
- Hojatolla Nikravesh
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, Faculty of Pharmacy, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Department of Toxicology, Faculty of Pharmacy, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Toxicology Research Center,Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Babak Behmanesh
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Department of Toxicology, Faculty of Pharmacy, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. .,Toxicology Research Center,Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Ali Teimoori
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Soheila Alboghobeish
- Department of Pharmacology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Zeidooni
- Department of Toxicology, Faculty of Pharmacy, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
16
|
Korsakova L, Krasko JA, Stankevicius E. Metabolic-targeted Combination Therapy With Dichloroacetate and Metformin Suppresses Glioblastoma Cell Line Growth In Vitro and In Vivo. In Vivo 2021; 35:341-348. [PMID: 33402483 DOI: 10.21873/invivo.12265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM We investigated the hypothesis that dichloroacetate (DCA), a pyruvate dehydrogenase kinase inhibitor, and metformin (MET), an antidiabetic agent and complex I inhibitor, have synergistic cytotoxic effects in glioblastoma cells in vitro and in vivo. MATERIALS AND METHODS We performed dose response experiments and combination index calculation. Apoptotic and necrotic cells were estimated by flow cytometry. Cell metabolism was evaluated by Seahorse analysis and lactate export. Overall survival and tumor volume growth experiments were performed in C57BL/6 mice GL-261 allograft model. RESULTS DCA and MET showed dose-dependent cytotoxicity and synergistic effects. DCA alleviated the increase in lactate production induced by MET. Seahorse analysis showed that DCA treatment results in increased oxygen consumption rate, which is decreased by MET. DCA and MET significantly inhibited tumor growth and increased overall survival in mice. CONCLUSION Compounds targeting tumor cell metabolism could become potential treatment options for glioblastoma multiforme.
Collapse
Affiliation(s)
- Laura Korsakova
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania;
| | | | - Edgaras Stankevicius
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania;
| |
Collapse
|
17
|
Malik P, Hoidal JR, Mukherjee TK. Recent Advances in Curcumin Treated Non-Small Cell Lung Cancers: An Impetus of Pleiotropic Traits and Nanocarrier Aided Delive ry. Curr Med Chem 2021; 28:3061-3106. [PMID: 32838707 DOI: 10.2174/0929867327666200824110332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 01/10/2023]
Abstract
Characterized by the abysmal 18% five year survival chances, non-small cell lung cancers (NSCLCs) claim more than half of their sufferers within the first year of being diagnosed. Advances in biomedical engineering and molecular characterization have reduced the NSCLC diagnosis via timid screening of altered gene expressions and impaired cellular responses. While targeted chemotherapy remains a major option for NSCLCs complications, delayed diagnosis, and concurrent multi-drug resistance remain potent hurdles in regaining normalcy, ultimately resulting in relapse. Curcumin administration presents a benign resolve herein, via simultaneous interception of distinctly expressed pathological markers through its pleiotropic attributes and enhanced tumor cell internalization of chemotherapeutic drugs. Studies on NSCLC cell lines and related xenograft models have revealed a consistent decline in tumor progression owing to enhanced chemotherapeutics cellular internalization via co-delivery with curcumin. This presents an optimum readiness for screening the corresponding effectiveness in clinical subjects. Curcumin is delivered to NSCLC cells either (i) alone, (ii) in stoichiometrically optimal combination with chemotherapeutic drugs, (iii) through nanocarriers, and (iv) nanocarrier co-delivered curcumin and chemotherapeutic drugs. Nanocarriers protect the encapsulated drug from accidental and non-specific spillage. A unanimous trait of all nanocarriers is their moderate drug-interactions, whereby native structural expressions are not tampered. With such insights, this article focuses on the implicit NSCLC curative mechanisms viz-a-viz, free curcumin, nanocarrier delivered curcumin, curcumin + chemotherapeutic drug and nanocarrier assisted curcumin + chemotherapeutic drug delivery.
Collapse
Affiliation(s)
- Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar, India
| | - John R Hoidal
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Tapan K Mukherjee
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
18
|
Synthesis and cytotoxic activity of organotin(IV) diallyldithiocarbamate compounds as anticancer agent towards colon adenocarcinoma cells (HT-29). Saudi J Biol Sci 2021; 28:3160-3168. [PMID: 34025187 PMCID: PMC8117248 DOI: 10.1016/j.sjbs.2021.02.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 01/08/2023] Open
Abstract
Context Diphenyltin(IV) diallyldithiocarbamate compound (Compound 1) and triphenyltin(IV) diallyldithiocarbamate compound (Compound 2) are two newly synthesised compounds of organotin(IV) with diallyldithiocarbamate ligands. Objective To assess the cytotoxic effects of two synthesised compounds against HT-29 human colon adenocarcinoma cells and human CCD-18Co normal colon cells. Materials and methods Two successfully synthesised compounds were characterised using elemental (carbon, hydrogen, nitrogen, and sulphur) analysis, Fourier-Transform Infrared (FTIR), and 1H, 13C 119Sn Nucleus Magnetic Resonance (NMR) spectroscopies. The single-crystal structure of both compounds was determined by X-ray single-crystal analysis. The cytotoxicity of the compounds was assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazholium bromide (MTT) assay upon 24 h of treatment. While the mode of cell death was determined based on the externalisation of phosphatidylserine using a flow cytometer. Results The elemental analysis data of the two compounds showed an agreement with the suggested formula of (C6H5)2Sn[S2CN(C3H5)2]2 for Compound 1 and (C6H5)3Sn[S2CN(C3H5)2] for Compound 2. The two major peaks of infrared absorbance, i.e., ν(C = N) and ν(C = S) were detected at the range of 1475–1479 cm−1 and 972–977 cm−1, respectively. The chemical shift of carbon in NCS2 group for Compound 1 and 2 were found at 200.82 and 197.79 ppm. The crystal structure of Compound 1 showed that it is six coordinated and crystallised in monoclinic, P21/c space group. While the crystal structure of Compound 2 is five coordinated and crystallised in monoclinic, P21/c space group. The cytotoxicity (IC50) of the two compounds against HT-29 cell were 2.36 μM and 0.39 μM. Meanwhile, the percentage of cell death modes between 60% and 75% for compound 1 and compound 2 were mainly due to apoptosis, suggesting that both compounds induced growth arrest. Conclusion Our study concluded that the synthesised compounds showed potent cytotoxicity towards HT-29 cell, with the triphenyltin(IV) compound showing the highest effect compared to diphenyltin(IV).
Collapse
|
19
|
Li J, Eu JQ, Kong LR, Wang L, Lim YC, Goh BC, Wong ALA. Targeting Metabolism in Cancer Cells and the Tumour Microenvironment for Cancer Therapy. Molecules 2020; 25:molecules25204831. [PMID: 33092283 PMCID: PMC7588013 DOI: 10.3390/molecules25204831] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Targeting altered tumour metabolism is an emerging therapeutic strategy for cancer treatment. The metabolic reprogramming that accompanies the development of malignancy creates targetable differences between cancer cells and normal cells, which may be exploited for therapy. There is also emerging evidence regarding the role of stromal components, creating an intricate metabolic network consisting of cancer cells, cancer-associated fibroblasts, endothelial cells, immune cells, and cancer stem cells. This metabolic rewiring and crosstalk with the tumour microenvironment play a key role in cell proliferation, metastasis, and the development of treatment resistance. In this review, we will discuss therapeutic opportunities, which arise from dysregulated metabolism and metabolic crosstalk, highlighting strategies that may aid in the precision targeting of altered tumour metabolism with a focus on combinatorial therapeutic strategies.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK;
| | - Jie Qing Eu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
| | - Li Ren Kong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Yaw Chyn Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pathology, National University Health System, Singapore 119074, Singapore
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Haematology-Oncology, National University Health System, Singapore 119228, Singapore
| | - Andrea L. A. Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; (J.Q.E.); (L.R.K.); (L.W.); (Y.C.L.); (B.C.G.)
- Department of Haematology-Oncology, National University Health System, Singapore 119228, Singapore
- Correspondence: ; Tel.: +65-6779-5555
| |
Collapse
|
20
|
Abánades Lázaro I, Wells CJR, Forgan RS. Multivariate Modulation of the Zr MOF UiO-66 for Defect-Controlled Combination Anticancer Drug Delivery. Angew Chem Int Ed Engl 2020; 59:5211-5217. [PMID: 31950568 PMCID: PMC7154787 DOI: 10.1002/anie.201915848] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Indexed: 01/05/2023]
Abstract
Metal-organic frameworks (MOFs) are emerging as leading candidates for nanoscale drug delivery, as a consequence of their high drug capacities, ease of functionality, and the ability to carefully engineer key physical properties. Despite many anticancer treatment regimens consisting of a cocktail of different drugs, examples of delivery of multiple drugs from one MOF are rare, potentially hampered by difficulties in postsynthetic loading of more than one cargo molecule. Herein, we report a new strategy, multivariate modulation, which allows incorporation of up to three drugs in the Zr MOF UiO-66 by defect-loading. The drugs are added to one-pot solvothermal synthesis and are distributed throughout the MOF at defect sites by coordination to the metal clusters. This tight binding comes with retention of crystallinity and porosity, allowing a fourth drug to be postsynthetically loaded into the MOFs to yield nanoparticles loaded with cocktails of drugs that show enhancements in selective anticancer cytotoxicity against MCF-7 breast cancer cells in vitro. We believe that multivariate modulation is a significant advance in the application of MOFs in biomedicine, and anticipate the protocol will also be adopted in other areas of MOF chemistry, to easily produce defective MOFs with arrays of highly functionalised pores for potential application in gas separations and catalysis.
Collapse
Affiliation(s)
- Isabel Abánades Lázaro
- WestCHEM School of ChemistryUniversity of GlasgowJoseph Black BuildingUniversity AvenueGlasgowG12 8QQUK
| | - Connor J. R. Wells
- WestCHEM School of ChemistryUniversity of GlasgowJoseph Black BuildingUniversity AvenueGlasgowG12 8QQUK
| | - Ross S. Forgan
- WestCHEM School of ChemistryUniversity of GlasgowJoseph Black BuildingUniversity AvenueGlasgowG12 8QQUK
| |
Collapse
|
21
|
Abánades Lázaro I, Wells CJR, Forgan RS. Multivariate Modulation of the Zr MOF UiO‐66 for Defect‐Controlled Combination Anticancer Drug Delivery. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201915848] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Isabel Abánades Lázaro
- WestCHEM School of ChemistryUniversity of GlasgowJoseph Black Building University Avenue Glasgow G12 8QQ UK
| | - Connor J. R. Wells
- WestCHEM School of ChemistryUniversity of GlasgowJoseph Black Building University Avenue Glasgow G12 8QQ UK
| | - Ross S. Forgan
- WestCHEM School of ChemistryUniversity of GlasgowJoseph Black Building University Avenue Glasgow G12 8QQ UK
| |
Collapse
|
22
|
Park SM, Jeon SK, Kim OH, Ahn JY, Kim CH, Park SD, Lee JH. Anti-tumor effects of the ethanolic extract of Trichosanthes kirilowii seeds in colorectal cancer. Chin Med 2019; 14:43. [PMID: 31624493 PMCID: PMC6781338 DOI: 10.1186/s13020-019-0263-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/19/2019] [Indexed: 01/16/2023] Open
Abstract
Background Trichosanthis semen, the seeds of Trichosanthes kirilowii Maxim. or Trichosanthes rosthornii Harms, has long been used in Korean medicine to loosen bowels and relieve chronic constipation. Although the fruits and radixes of this medicinal herb and their constituents have been reported to exhibit therapeutic effects in various cancers, the anti-cancer effects of its seeds have been relatively less studied. In this study, we investigated the effects of an ethanolic extract of T. kirilowii seeds (TKSE) against colorectal cancer and its mechanism. Methods The anti-tumor effects of the TKSE were evaluated in HT-29 and CT-26 colorectal cancer cells and in a CT-26 tumor-bearing mouse model. Results TKSE suppressed the growth of HT-29 and CT-26 cells (both colorectal cancer cell lines) and the cytotoxic effect of TKSE was greater than that of 5-fluorouracil (5-Fu) in HT-29 cells. TKSE significantly induced mitochondrial membrane potential loss in HT-29 and CT-26 cells and dose-dependently inhibited Bcl-2 expression and induced the cleavages of caspase-3 and PARP. In particular, TKSE at 300 µg/mL induced nuclear condensation and fragmentation in HT-29 cells. Furthermore, TKSE dose-dependently inhibited activations of the Akt/mTOR and ERK pathways, and markedly induced the phosphorylation of AMPK. An AMPKα inhibitor (compound C) effectively blocked the TKSE-induced mitochondrial dysfunction. In addition, TKSE attenuated the hypoxia-inducible factor-1α/vascular endothelial growth factor signaling pathway in HT-29 cells under hypoxic-mimic conditions and inhibited migration and invasion. Oral administration of TKSE (100 or 300 mg/kg) inhibited tumor growth in a mouse CT-26 allograft model but was not as effective as 5-Fu (the positive control), which was administered intraperitoneally. In the same model, 5-Fu caused significant body weight loss, but no such loss was observed in TKSE-treated mice. Conclusion Taken together, these results suggest TKSE has potent anti-tumor effects which might be partly due to the activation of AMPK, and the induction mitochondrial-mediated apoptosis in colorectal cancer cells. These findings provide scientific evidence supporting the potential use of TKSE as a complementary and alternative medicine for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Su Mi Park
- 1Department of Korean Medicine, College of Korean Medicine, Dongguk University, Goyang, 10326 Republic of Korea
| | - Sang Kyu Jeon
- 1Department of Korean Medicine, College of Korean Medicine, Dongguk University, Goyang, 10326 Republic of Korea
| | - Ok Hyeon Kim
- 1Department of Korean Medicine, College of Korean Medicine, Dongguk University, Goyang, 10326 Republic of Korea
| | - Jung Yun Ahn
- 1Department of Korean Medicine, College of Korean Medicine, Dongguk University, Goyang, 10326 Republic of Korea
| | - Chang-Hyun Kim
- 2Department of Medicine, College of Medicine, Dongguk University, Goyang, 10326 Republic of Korea
| | - Sun-Dong Park
- 1Department of Korean Medicine, College of Korean Medicine, Dongguk University, Goyang, 10326 Republic of Korea
| | - Ju-Hee Lee
- 1Department of Korean Medicine, College of Korean Medicine, Dongguk University, Goyang, 10326 Republic of Korea
| |
Collapse
|
23
|
Kim KT, Angerani S, Chang D, Winssinger N. Coupling of DNA Circuit and Templated Reactions for Quadratic Amplification and Release of Functional Molecules. J Am Chem Soc 2019; 141:16288-16295. [DOI: 10.1021/jacs.9b05688] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ki Tae Kim
- Department of Organic Chemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 30 quai Ernest Ansermet, 1205 Geneva, Switzerland
| | - Simona Angerani
- Department of Organic Chemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 30 quai Ernest Ansermet, 1205 Geneva, Switzerland
| | - Dalu Chang
- Department of Organic Chemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 30 quai Ernest Ansermet, 1205 Geneva, Switzerland
| | - Nicolas Winssinger
- Department of Organic Chemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 30 quai Ernest Ansermet, 1205 Geneva, Switzerland
| |
Collapse
|
24
|
Ludman T, Melemedjian OK. Bortezomib-induced aerobic glycolysis contributes to chemotherapy-induced painful peripheral neuropathy. Mol Pain 2019; 15:1744806919837429. [PMID: 30810076 PMCID: PMC6452581 DOI: 10.1177/1744806919837429] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Chemotherapy-induced painful peripheral neuropathy (CIPN) is the most common toxicity associated with widely used chemotherapeutics. CIPN is the major cause of dose reduction or discontinuation of otherwise life-saving treatment. Unfortunately, CIPN can persist in cancer survivors, which adversely affects their quality of life. Moreover, available treatments are vastly inadequate, warranting a better understanding of the biochemical and metabolic mechanisms that occur in response to chemotherapeutics which would be critical for the development of novel therapies for CIPN. Using extracellular flux analysis, this study demonstrated that the proteasome inhibitor, bortezomib, enhanced glycolysis while suppressing oxidative phosphorylation in the sensory neurons of mice. This metabolic phenotype is known as aerobic glycolysis. Bortezomib upregulated lactate dehydrogenase A and pyruvate dehydrogenase kinase 1, which consequently enhanced the production of lactate and repressed pyruvate oxidation, respectively. Moreover, lactate dehydrogenase A- and pyruvate dehydrogenase kinase 1-driven aerobic glycolysis was associated with increased extracellular acidification, augmented calcium responses, and pain in bortezomib-induced CIPN. Remarkably, pharmacological blockade and in vivo knockdown of lactate dehydrogenase A or pyruvate dehydrogenase kinase 1 reversed the metabolic phenotype, attenuated calcium responses, and alleviated pain induced by bortezomib. Collectively, these results elucidate the mechanisms by which bortezomib induces aerobic glycolysis. Moreover, these findings establish aerobic glycolysis as a metabolic phenotype that underpins bortezomib-induced CIPN.
Collapse
Affiliation(s)
- Taylor Ludman
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD, USA
| | - Ohannes K Melemedjian
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD, USA.,2 Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, USA
| |
Collapse
|
25
|
Kumar B, Murali A, Bharath AB, Giri S. Guar gum modified upconversion nanocomposites for colorectal cancer treatment through enzyme-responsive drug release and NIR-triggered photodynamic therapy. NANOTECHNOLOGY 2019; 30:315102. [PMID: 30893650 DOI: 10.1088/1361-6528/ab116e] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Multimodal therapeutic approach towards colorectal cancer (CRC) holds great promise. There is, however, no convincing strategy reported to date that employs a multimodal strategy in CRC treatment. The present study reports an intense green-emitting core-shell photoluminescent upconversion (CSGU) nanocrystal engineered to synergistically perform photodynamic and enzyme-triggered delivery of the chemotherapeutic agent for an enhanced therapeutic outcome on HT-29 colon carcinoma cells in vitro. The photodynamic activity is achieved by the energy transfer between CSGU and the chemically conjugated Rose Bengal (RB) molecules that are further protected by a mesoporous silica (MS) layer. The chemical assay demonstrates a remarkable FRET mediated generation of 1O2 under NIR (980 nm) excitation. The outermost MS layer of the nanoplatform is utilized for the loading of the 5FU anticancer drug, which is further capped with a guar gum (GG) polysaccharide polymer. The release of the 5FU is specifically triggered by the degradation of the GG cap by specific enzymes secreted from colonic microflora, which otherwise showed 'zero-release behavior' in the absence of any enzymatic trigger in various simulated gastro-intestinal (GI) conditions. Furthermore, the enhanced therapeutic efficacy of the nanoplatform (CSGUR-MSGG/5FU) was evaluated through in vitro studies using HT-29 CRC cell lines by various biochemical and microscopic assays by the simultaneous triggering effect of colonic enzyme and 980 nm laser excitation. In addition, the strong visible emission from the nanoplatform has been utilized for NIR-induced cellular bioimaging.
Collapse
Affiliation(s)
- Balmiki Kumar
- Department of Chemistry, National Institute of Technology, Rourkela. Odisha-769008, India
| | | | | | | |
Collapse
|
26
|
Sun L, Xu Y, Gao Y, Huang X, Feng S, Chen J, Wang X, Guo L, Li M, Meng X, Zhang J, Ge J, An X, Ding D, Luo Y, Zhang Y, Jiang Q, Ning X. Synergistic Amplification of Oxidative Stress-Mediated Antitumor Activity via Liposomal Dichloroacetic Acid and MOF-Fe 2. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1901156. [PMID: 31074196 DOI: 10.1002/smll.201901156] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/21/2019] [Indexed: 06/09/2023]
Abstract
Cancer cells are susceptible to oxidative stress; therefore, selective elevation of intracellular reactive oxygen species (ROS) is considered as an effective antitumor treatment. Here, a liposomal formulation of dichloroacetic acid (DCA) and metal-organic framework (MOF)-Fe2+ (MD@Lip) has been developed, which can efficiently stimulate ROS-mediated cancer cell apoptosis in vitro and in vivo. MD@Lip can not only improve aqueous solubility of octahedral MOF-Fe2+ , but also generate an acidic microenvironment to activate a MOF-Fe2+ -based Fenton reaction. Importantly, MD@Lip promotes DCA-mediated mitochondrial aerobic oxidation to increase intracellular hydrogen peroxide (H2 O2 ), which can be consequently converted to highly cytotoxic hydroxyl radicals (•OH) via MOF-Fe2+ , leading to amplification of cancer cell apoptosis. Particularly, MD@Lip can selectively accumulate in tumors, and efficiently inhibit tumor growth with minimal systemic adverse effects. Therefore, liposome-based combination therapy of DCA and MOF-Fe2+ provides a promising oxidative stress-associated antitumor strategy for the management of malignant tumors.
Collapse
Affiliation(s)
- Lei Sun
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Ya Gao
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Xinyu Huang
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Shujun Feng
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmei Chen
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Xuekun Wang
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Leilei Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, Center of Advanced Pharmaceutics and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Meng Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
| | - Xia Meng
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Jikang Zhang
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Junliang Ge
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Xueying An
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China
| | - Dang Ding
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Yadong Luo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
| | - Yu Zhang
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
27
|
Sun T, Zhao Q, Zhang C, Cao L, Song M, Maimela NR, Liu S, Wang J, Gao Q, Qin G, Wang L, Zhang Y. Screening common signaling pathways associated with drug resistance in non-small cell lung cancer via gene expression profile analysis. Cancer Med 2019; 8:3059-3071. [PMID: 31025554 PMCID: PMC6558586 DOI: 10.1002/cam4.2190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Although several therapeutic strategies have been employed to curb lung cancer, the survival rate is still poor owing to the development of drug resistance. The mechanisms underlying drug resistance development are incompletely understood. Here, we aimed to identify the common signaling pathways involved in drug resistance in non-small cell lung cancer (NSCLC). Three published transcriptome microarray data were downloaded from the Gene Expression Omnibus (GEO) database comprising different drug-resistant cell lines and their parental cell lines. Differentially expressed genes (DEGs) were identified and used to perform Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. An overlapping analysis was performed for KEGG pathways enriched from all the three datasets to identify the common signaling pathways. As a result, we found that metabolic pathways, ubiquitin-mediated proteolysis, and mitogen-activated protein kinase (MAPK) signaling were the most aberrantly expressed signaling pathways. The knockdown of nicotinamide phosphoribosyltransferase (NAMPT), the gene involved in metabolic pathways and known to be upregulated in drug-resistant tumor cells, was shown to increase the apoptosis of cisplatin-resistant A549 cells following cisplatin treatment. Thus, our results provide an in-depth analysis of the signaling pathways that are commonly altered in drug-resistant NSCLC cell lines and highlight the potential strategy that facilitates the development of interventions to interfere with upregulated signaling pathways as well as to boost downregulated signaling pathways in drug-resistant tumors for the elimination of multiple resistance of NSCLC.
Collapse
Affiliation(s)
- Ting Sun
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Department of Respiratory medicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Qitai Zhao
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Cancer CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chaoqi Zhang
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Cancer CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ling Cao
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Mengjia Song
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | | | - Shasha Liu
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jinjin Wang
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Qun Gao
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Cancer CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guohui Qin
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Liping Wang
- Cancer CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yi Zhang
- Biotherapy CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Cancer CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
- Engineering Key Laboratory for Cell Therapy of Henan ProvinceZhengzhouChina
| |
Collapse
|
28
|
Combinatorial Cytotoxic Effects of Gelam Honey and 5-Fluorouracil against Human Adenocarcinoma Colon Cancer HT-29 Cells In Vitro. Int J Cell Biol 2019; 2019:3059687. [PMID: 30923553 PMCID: PMC6408991 DOI: 10.1155/2019/3059687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/09/2019] [Accepted: 01/17/2019] [Indexed: 11/18/2022] Open
Abstract
Combination of natural products with chemodrugs is becoming a trend in discovering new therapeutics approach for enhancing the cancer treatment process. In the present study, we aimed to investigate the cytotoxic and apoptosis induction of Gelam honey (GH) combined with or without 5-Fluorouracil (5-FU) on HT-29 cells. The cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay to assess cytotoxicity. Morphological changes and apoptosis were determined by the inverted microscope, Annexin V-FITC, and DNA fragmentation via flow cytometric analysis, respectively. Our results demonstrate that combined treatment revealed a remarkable and concentration-dependent cytotoxic effect on HT-29 cells in comparison with GH and 5-FU alone. Flow cytometry analysis showed that early apoptosis event was more pronounced in combined treatment. In addition, compared to 5-FU alone, apoptosis of HT-29 cells treated with combinations of GH and 5-FU demonstrated increasing percentages of fragmented DNA. Our results suggest that GH has a synergistic cytotoxic effect with 5-FU in HT-29 cell lines in vitro. Although the actions of the molecular mechanisms are not yet clear, the results reveal that the combination of GH and 5-FU could have the potential as a therapeutic agent.
Collapse
|
29
|
Abánades Lázaro I, Forgan RS. Application of zirconium MOFs in drug delivery and biomedicine. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2018.09.009] [Citation(s) in RCA: 331] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
30
|
Palamarciuc O, Milunović MNM, Sîrbu A, Stratulat E, Pui A, Gligorijevic N, Radulovic S, Kožíšek J, Darvasiová D, Rapta P, Enyedy EA, Novitchi G, Shova S, Arion VB. Investigation of the cytotoxic potential of methyl imidazole-derived thiosemicarbazones and their copper(ii) complexes with dichloroacetate as a co-ligand. NEW J CHEM 2019. [DOI: 10.1039/c8nj04041a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Investigation of the cytotoxic potential of imidazole-derived thiosemicarbazones and their copper(ii) complexes with CHCl2CO2− as a co-ligand.
Collapse
|
31
|
El Sayed SM, Baghdadi H, Ahmed NS, Almaramhy HH, Mahmoud AAA, El-Sawy SA, Ayat M, Elshazley M, Abdel-Aziz W, Abdel-Latif HM, Ibrahim W, Aboonq MS. Dichloroacetate is an antimetabolite that antagonizes acetate and deprives cancer cells from its benefits: A novel evidence-based medical hypothesis. Med Hypotheses 2019; 122:206-209. [PMID: 30593413 DOI: 10.1016/j.mehy.2018.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/28/2018] [Accepted: 11/20/2018] [Indexed: 01/22/2023]
Abstract
Dichloroacetate (DCA) is a promising safe anticancer drug that cured a patient with chemoresistant non-Hodgkin's lymphoma and treated lactic acidosis effectively. The well-known mechanism of DCA action is through stimulating Krebs cycle (stimulating pyruvate dehydrogenase via inhibiting pyruvate dehydrogenase kinase). This prevents lactate formation (Warburg effect) depriving cancer cells of lactate-based benefits e.g. angiogenesis, chemoresistance and radioresistance. Here, we introduce novel evidence-based hypotheses to explain DCA-induced anticancer effects. On pharmacological and biochemical bases, we hypothesize that DCA is a structural antagonist of acetate competing with it for target enzymes and biological reactions. We hypothesize that DCA exerts its anticancer effects via depriving cancer of acetate benefits. We hypothesize also that acetate is an antidote of DCA capable of treating DCA toxicity. Many reports support our hypotheses. Acetate is vital for cancer cells (tumors depend on acetate) and DCA is structurally similar to acetate. DCA exerts opposite effects to acetate. Acetate caused a decrease in serum potassium, phosphorus and glucose, and an increase in serum lactate, citrate, free fatty acids and ketone bodies (serum acetoacetate and beta-hydroxybutyrate levels). Acetate decreased the proportion of active (dephosphorylated) pyruvate dehydrogenase in perfused rat heart. DCA produced quite opposite effects. Intravenous infusion of acetate produced metabolic alkalemia while DCA caused minimal effects on acid-base status. Acetate is important for cancer cells metabolism and survival as elevated acetate can drive resistance to targeted cancer treatments. Acetate is required for epidermal growth factor receptor vIII mutation in lethal brain tumors. Experimentally, DCA inhibited acetate oxidation in hearts of normal rats and reversed inhibitory effects of acetate on the oxidation of glucose. During presence of DCA with no glucose in heart perfusions with [1-14C]acetate, DCA decreased the specific radioactivity of acetyl CoA and its product citrate. This proves our hypotheses that DCA is an antimetabolite that antagonizes acetate for vital reactions in cancer cells. Acetate may be used as an antidote to combat DCA toxicity.
Collapse
Affiliation(s)
- Salah Mohamed El Sayed
- Department of Clinical Biochemistry and Molecular Medicine, Taibah College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia; Department of Medical Biochemistry, Sohag Faculty of Medicine, Sohag University, Egypt.
| | - Hussam Baghdadi
- Department of Clinical Biochemistry and Molecular Medicine, Taibah College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| | - Nagwa Sayed Ahmed
- Department of Medical Biochemistry, Sohag Faculty of Medicine, Sohag University, Egypt
| | - Hamdi H Almaramhy
- Department of Surgery, Taibah College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| | - Ahmed Al-Amir Mahmoud
- Department of Medical Biochemistry, Sohag Faculty of Medicine, Sohag University, Egypt
| | - Samer Ahmed El-Sawy
- Department of Medical Biochemistry, Sohag Faculty of Medicine, Sohag University, Egypt
| | - Mongi Ayat
- Department of Clinical Biochemistry and Molecular Medicine, Taibah College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| | - Momen Elshazley
- Department of Medicine, Taibah College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia; Department of Occupational Diseases and Toxigenomics, Sohag Faculty of Medicine, Sohag University, Egypt
| | - Wafaa Abdel-Aziz
- Department of Medical Pharmacology, Sohag Faculty of Medicine, Sohag University, Egypt
| | | | - Walaa Ibrahim
- Department of Medical Pharmacology, Sohag Faculty of Medicine, Sohag University, Egypt
| | - Moutasem Salih Aboonq
- Department of Physiology, Taibah College of Medicine, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| |
Collapse
|
32
|
Dichloroacetate and Salinomycin Exert a Synergistic Cytotoxic Effect in Colorectal Cancer Cell Lines. Sci Rep 2018; 8:17744. [PMID: 30531808 PMCID: PMC6288092 DOI: 10.1038/s41598-018-35815-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022] Open
Abstract
In the present study, we examined a hypothesis that dichloroacetate, a metabolic inhibitor, might efficiently potentiate the cytotoxic effect of salinomycin, an antibiotic ionophore, on two human colorectal cancer derived cell lines DLD-1 and HCT116. First, we performed a series of dose response experiments in the 2D cell culture by applying mono- and combination therapy and by using the Chou-Talalay method found that salinomycin in combination with dichloroacetate acted synergistically in both cell lines. Secondly, in order to recapitulate the in vivo tumor architecture, we tested various doses of these compounds, alone and in combination, in the 3D multicellular spheroid culture. The effect of combination of dichloracetate and salinomycin on multicellular spheroid size was stronger than the sum of both monotherapies, particularly in HCT116 cells. Further, we demonstrate that the synergistic effect of compounds may be related to the inhibitory effect of dichloroacetate on multidrug resistance proteins, and in contrast, it is not related to dichloroacetate-induced reduction of intracellular pH. Our findings indicate that the combination therapy of salinomycin and dichloroacetate could be an effective option for colorectal cancer treatment and provide the first mechanistic explanation of the synergistic action of these compounds.
Collapse
|
33
|
Abánades Lázaro I, Haddad S, Rodrigo-Muñoz JM, Marshall RJ, Sastre B, Del Pozo V, Fairen-Jimenez D, Forgan RS. Surface-Functionalization of Zr-Fumarate MOF for Selective Cytotoxicity and Immune System Compatibility in Nanoscale Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:31146-31157. [PMID: 30136840 DOI: 10.1021/acsami.8b11652] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Metal-organic frameworks (MOFs), network structures wherein metal ions or clusters link organic ligands into porous materials, are being actively researched as nanoscale drug delivery devices as they offer tunable structures with high cargo loading that can easily be further functionalized for targeting and enhanced physiological stability. The excellent biocompatibility of Zr has meant that its MOFs are among the most studied to date, in particular the archetypal Zr terephthalate UiO-66. In contrast, the isoreticular analog linked by fumarate (Zr-fum) has received little attention, despite the endogenous linker being part of the Krebs cycle. Herein, we report a comprehensive study of Zr-fum in the context of drug delivery. Reducing particle size is shown to increase uptake by cancer cells while reducing internalization by macrophages, immune system cells that remove foreign objects from the bloodstream. Zr-fum is compatible with defect loading of the drug dichloroacetate (DCA) as well as surface modification during synthesis, through coordination modulation and postsynthetically. DCA-loaded, PEGylated Zr-fum shows selective in vitro cytotoxicity toward HeLa and MCF-7 cancer cells, likely as a consequence of its enhanced caveolae-mediated endocytosis compared to uncoated precursors, and it is well tolerated by HEK293 kidney cells, J774 macrophages, and human peripheral blood lymphocytes. Compared to UiO-66, Zr-fum is more efficient at transporting the drug mimic calcein into HeLa cells, and DCA-loaded, PEGylated Zr-fum is more effective at reducing HeLa and MCF-7 cell proliferation than the analogous UiO-66 sample. In vitro examination of immune system response shows that Zr-fum samples induce less reactive oxygen species than UiO-66 analogs, possibly as a consequence of the linker being endogenous, and do not activate the C3 and C4 complement cascade pathways, suggesting that Zr-fum can avoid phagocytic activation. The results show that Zr-fum is an attractive alternative to UiO-66 for nanoscale drug delivery, and that a wide range of in vitro experiments is available to greatly inform the design of drug delivery systems prior to early stage animal studies.
Collapse
Affiliation(s)
- Isabel Abánades Lázaro
- WestCHEM School of Chemistry , University of Glasgow , Joseph Black Building, University Avenue , Glasgow G12 8QQ , U.K
| | - Salame Haddad
- Adsorption & Advanced Materials Laboratory, Department of Chemical Engineering & Biotechnology , University of Cambridge , Philippa Fawcett Drive , Cambridge CB3 0AS , U.K
| | - Jose M Rodrigo-Muñoz
- Department of Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz , Universidad Autónoma de Madrid (IIS-FJD, UAM), and CIBER de Enfermedades Respiratorias (CIBERES) , 28029 Madrid , Spain
| | - Ross J Marshall
- WestCHEM School of Chemistry , University of Glasgow , Joseph Black Building, University Avenue , Glasgow G12 8QQ , U.K
| | - Beatriz Sastre
- Department of Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz , Universidad Autónoma de Madrid (IIS-FJD, UAM), and CIBER de Enfermedades Respiratorias (CIBERES) , 28029 Madrid , Spain
| | - Victoria Del Pozo
- Department of Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz , Universidad Autónoma de Madrid (IIS-FJD, UAM), and CIBER de Enfermedades Respiratorias (CIBERES) , 28029 Madrid , Spain
| | - David Fairen-Jimenez
- Adsorption & Advanced Materials Laboratory, Department of Chemical Engineering & Biotechnology , University of Cambridge , Philippa Fawcett Drive , Cambridge CB3 0AS , U.K
| | - Ross S Forgan
- WestCHEM School of Chemistry , University of Glasgow , Joseph Black Building, University Avenue , Glasgow G12 8QQ , U.K
| |
Collapse
|
34
|
Li B, Zhu Y, Sun Q, Yu C, Chen L, Tian Y, Yan J. Reversal of the Warburg effect with DCA in PDGF‑treated human PASMC is potentiated by pyruvate dehydrogenase kinase‑1 inhibition mediated through blocking Akt/GSK‑3β signalling. Int J Mol Med 2018; 42:1391-1400. [PMID: 29956736 PMCID: PMC6089770 DOI: 10.3892/ijmm.2018.3745] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/25/2018] [Indexed: 01/12/2023] Open
Abstract
There is accumulating evidence indicating that the growth inhibitory effect of dichloroacetate (DCA) on pulmonary arterial smooth muscle cells (PASMCs) may be associated with the reversal of the Warburg effect and initiation of the mitochondria‑dependent apoptotic pathway. Previous studies indicated that platelet‑derived growth factor (PDGF) promoted the Warburg effect and resulted in apoptotic resistance of PASMCs, which was attributed to activation of the phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (Akt) signalling pathway. However, the mechanism underlying the pro‑apoptotic effect of DCA on PDGF‑treated PASMCs has not been thoroughly elucidated, and the effect of the Akt/glycogen synthase kinase‑3β (GSK‑3β) pathway inhibition concomitant with the effect of DCA on PASMC proliferation remains unclear. The growth of human PASMCs and the lactate concentration in extracellular medium of PASMCs were detected by Cell Counting Kit‑8 assays and a Lactate Colorimetric Assay kit, respectively. Cell apoptosis was evaluated by fluorescence activated cell sorting. The mitochondrial membrane potential (ΔΨm) was assessed with 5,5',6,6'‑tetrachloro‑1,1',3,3'‑tetraethylbenzimidazol‑carbocyanine iodide assays. The expression levels of phosphorylated Akt and GSK‑3β, pyruvate dehydrogenase, cleaved caspase‑3, pyruvate dehydrogenase kinase‑1 (PDK‑1), hypoxia inducible factor‑1α (HIF‑1α) and hexokinase‑2 (HK‑2) were measured with western blot analysis. Confocal analyses were employed to determine HK‑2 co‑localisation with the mitochondria. The results indicated that DCA inhibited human PASMC proliferation in a dose‑dependent manner. DCA at 10 mM promoted apoptosis and the upregulation of activated caspase‑3 in PASMCs pre‑treated with 20 ng/ml PDGF‑homeodimer BB (BB). Treatment with 5 µM LY294002 produced minimal anti‑proliferative effects on human PASMCs and barely induced cellular apoptosis and caspase‑3 activation. However, co‑administration of 10 mM DCA with LY294002 significantly decreased the cell proliferation index and induced cell apoptosis and caspase‑3 activation. The combined administration of LY294002 with DCA significantly decreased lactate concentration, promoted the depolarisation of the ΔΨm and repressed HIF‑1α upregulation and HK‑2 activation in PASMCs treated with PDGF, which was attributed to the potentiation of DCA‑induced PDK‑1 inhibition by LY294002 via blockade of the Akt/GSK‑3β/HIF‑1α signalling pathway. In conclusion, inhibition of the Akt/GSK‑3β pathway improved the pro‑apoptotic effect of DCA on human PASMCs, which may be attributed to a reversal of the Warburg effect by blocking the mutual interaction between HIF‑1α and PDK‑1, consequently downregulating HK‑2. Therefore, combinatory treatment with DCA and PI3K inhibitors may represent a novel therapeutic strategy for the reversal of apoptosis resistance exhibited by PASMCs as a result of mitochondrial bioenergetic abnormalities, as well as the treatment of pulmonary vascular remodelling in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Bingbing Li
- Department of Anaesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008
| | - Yuling Zhu
- Department of Anaesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008
| | - Qing Sun
- Department of Anaesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008
| | - Chunfang Yu
- Department of Anaesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008
| | - Lian Chen
- Department of Anaesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008
| | - Yali Tian
- Department of Anaesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008
| | - Jie Yan
- Department of Anaesthesiology, The Affiliated Obstetrics and Gynaecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, P.R. China
| |
Collapse
|
35
|
Wang A, Wang M, Pang Q, Jia L, Zhao J, Chen M, Zhao Y. Lily extracts inhibit the proliferation of gastric carcinoma SGC-7901 cells by affecting cell cycle progression and apoptosis via the upregulation of caspase-3 and Fas proteins, and the downregulation of FasL protein. Oncol Lett 2018; 16:1397-1404. [PMID: 30008816 PMCID: PMC6036323 DOI: 10.3892/ol.2018.8806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 06/02/2017] [Indexed: 01/08/2023] Open
Abstract
The present study aimed to investigate the effect of alkaloids and carbinol extracts from lily on the proliferation of SGC-7901 cells, as well as the underlying mechanism. SGC-7901 cells were incubated with different concentrations of alkaloid or carbinol extracts for 24, 48 or 72 h. MTT assays were used to measure the inhibition rate of SGC-7901 cell proliferation. Inverted phase contrast and fluorescence microscopy was used to observe morphological changes of SGC-7901 cells. Flow cytometry was employed to detect cell cycle progression and apoptosis rates of SGC-7901 cells. Western blotting was performed to measure the expression of caspase-3, Fas and Fas ligand (FasL) proteins in SGC-7901 cells. The inhibition rate of SGC-7901 cell proliferation was significantly enhanced with increasing drug concentrations and time elapsed. Treatment with alkaloid or carbinol extracts deteriorated the morphology of SGC-7901 cells in a dose-dependent manner. Alkaloid and carbinol extracts arrested SGC-7901 cells in the G2/M phase, and induced apoptosis in a dose-dependent manner. Alkaloid and carbinol extracts enhanced caspase-3, and Fas expression, but reduced FasL expression in SGC-7901 cells. The present study demonstrated that alkaloids and carbinol extracts from lily inhibited the proliferation of gastric carcinoma SGC-7901 cells by arresting cells in the G2/M phase. The upregulation of caspase-3 and Fas proteins, and the downregulation of FasL protein may be an important mechanism for the induction of SGC-7901 cell apoptosis.
Collapse
Affiliation(s)
- Aihong Wang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Mingquan Wang
- Department of Interventional Radiology, Affiliated Hospital of Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Qiuxia Pang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Lei Jia
- Department of Laboratory Medicine, Fuxian County Hospital, Yan'an, Shaanxi 727500, P.R. China
| | - Jumei Zhao
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Meini Chen
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yufeng Zhao
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| |
Collapse
|
36
|
Kouidhi S, Ben Ayed F, Benammar Elgaaied A. Targeting Tumor Metabolism: A New Challenge to Improve Immunotherapy. Front Immunol 2018; 9:353. [PMID: 29527212 PMCID: PMC5829092 DOI: 10.3389/fimmu.2018.00353] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/07/2018] [Indexed: 12/22/2022] Open
Abstract
Currently, a marked number of clinical trials on cancer treatment have revealed the success of immunomodulatory therapies based on immune checkpoint inhibitors that activate tumor-specific T cells. However, the therapeutic efficacy of cancer immunotherapies is only restricted to a small fraction of patients. A deeper understanding of key mechanisms generating an immunosuppressive tumor microenvironment (TME) remains a major challenge for more effective antitumor immunity. There is a growing evidence that the TME supports inappropriate metabolic reprogramming that dampens T cell function, and therefore impacts the antitumor immune response and tumor progression. Notably, the immunosuppressive TME is characterized by a lack of crucial carbon sources critical for T cell function and increased inhibitory signals. Here, we summarize the basics of intrinsic and extrinsic metabolic remodeling and metabolic checkpoints underlying the competition between cancer and infiltrating immune cells for nutrients and metabolites. Intriguingly, the upregulation of tumor programmed death-L1 and cytotoxic T lymphocyte-associated antigen 4 alters the metabolic programme of T cells and drives their exhaustion. In this context, targeting both tumor and T cell metabolism can beneficially enhance or temper immunity in an inhospitable microenvironment and markedly improve the success of immunotherapies.
Collapse
Affiliation(s)
- Soumaya Kouidhi
- Laboratory BVBGR, LR11ES31, Higher Institute of Biotechnology of Sidi Thabet (ISBST), Department of Biotechnology, University of Manouba, Sidi Thabet, Tunisia
- Laboratory of Genetics, Immunology and Human Pathology, Faculty of Sciences of Tunis, Department of Biology, University Tunis El Manar, Tunis, Tunisia
| | - Farhat Ben Ayed
- Association Tunisienne de Lutte contre le Cancer (ATCC), Tunis, Tunisia
| | - Amel Benammar Elgaaied
- Laboratory of Genetics, Immunology and Human Pathology, Faculty of Sciences of Tunis, Department of Biology, University Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
37
|
A key role for transketolase-like 1 in tumor metabolic reprogramming. Oncotarget 2018; 7:51875-51897. [PMID: 27391434 PMCID: PMC5239521 DOI: 10.18632/oncotarget.10429] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/13/2016] [Indexed: 12/15/2022] Open
Abstract
Metabolic reprogramming, a crucial cancer hallmark, shifts metabolic pathways such as glycolysis, tricarboxylic acid cycle or lipogenesis, to enable the growth characteristics of cancer cells. Here, we provide evidence that transketolase-like 1 (TKTL1) orchestrates aerobic glycolysis, fatty acid and nucleic acid synthesis, glutamine metabolism, protection against oxidative stress and cell proliferation. Furthermore, silencing of TKTL1 reduced the levels of sphingolipids such as lactosylceramide (a sphingolipid regulating cell survival, proliferation and angiogenesis) and phosphatidylinositol (which activates PI3K/Akt/mTOR signaling). Thus, in addition to its well-known roles in glucose and amino acid metabolism, TKTL1 also regulates lipid metabolism. In conclusion, our study provides unprecedented evidence that TKTL1 plays central roles in major metabolic processes subject to reprogramming in cancer cells and thus identifies TKTL1 as a promising target for new anti-cancer therapies.
Collapse
|
38
|
Nunes RC, Ribeiro CJA, Monteiro Â, Rodrigues CMP, Amaral JD, Santos MMM. In vitro targeting of colon cancer cells using spiropyrazoline oxindoles. Eur J Med Chem 2017; 139:168-179. [PMID: 28800455 DOI: 10.1016/j.ejmech.2017.07.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/23/2017] [Accepted: 07/23/2017] [Indexed: 02/07/2023]
Abstract
We report on the synthesis and biological evaluation of a library of twenty-three spiropyrazoline oxindoles. The antiproliferative activity of the chemical library was evaluated in HCT-116 p53(+/+) human colon cancer cell line with eight derivatives displaying good activities (IC50<15 μM). To characterize the molecular mechanisms involved in compound antitumoral activity, two spiropyrazoline oxindoles were selected for further studies. Both compounds were able to induce apoptosis and cell cycle arrest at G0/G1 phase and upregulated p53 steady-state levels, while decreasing its main inhibitor MDM2. Importantly, cytotoxic effects induced by spiropyrazolines oxindoles occurred in cancer cells without eliciting cell death in non-malignant CCD-18Co human colon fibroblasts. Additionally, we demonstrated that the combination of spiropyrazoline oxindole 2e with sub-toxic concentrations of the chemotherapeutic agent 5-fluorouracil (5-FU) exerted a synergistic inhibitory effect on HCT-116 colon cancer cell proliferation. Collectively, our results show the potential of spiropyrazoline oxindoles for development of novel anticancer agents.
Collapse
Affiliation(s)
- Rute C Nunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Carlos J A Ribeiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Ângelo Monteiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Joana D Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| | - Maria M M Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal. /
| |
Collapse
|
39
|
Jia HY, Wang HN, Xia FY, Sun Y, Liu HL, Yan LL, Li SS, Jiang DC, Xu MM. Dichloroacetate induces protective autophagy in esophageal squamous carcinoma cells. Oncol Lett 2017; 14:2765-2770. [PMID: 28928817 PMCID: PMC5588168 DOI: 10.3892/ol.2017.6562] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/27/2017] [Indexed: 11/06/2022] Open
Abstract
Dichloroacetate (DCA) is an inhibitor of pyruvate dehydrogenase kinase, which promotes the flux of carbohydrates into mitochondria and enhances the aerobic oxidation of glucose. DCA has previously been demonstrated to exhibit antitumor properties. The present study revealed that treatment with DCA induced increased levels of autophagy-associated proteins in esophageal squamous carcinoma cells while minimally affecting apoptosis. The present study examined the localization of light chain (LC)-3 by adenovirus infection with a green fluorescent protein (FP)-red FP-LC3 reporter construction and confirmed that DCA treatment induced significant autophagy. Furthermore, the inhibition of DCA-induced autophagy facilitated cell apoptosis and improved the drug sensitivity of esophageal squamous carcinoma cells to DCA and 5-FU (5-fluorouracil). The proliferation of TE-1 cells was markedly inhibited at low concentrations of DCA and 5-FU treatment when subjected to Atg5 mRNA interference, indicating that autophagy performed a protective role in cell survival upon DCA treatment. To determine the underlying mechanism of DCA-induced autophagy, the present study measured alterations in autophagy-associated signaling pathways. Notably, the protein kinase B (Akt)-mechanistic target of rapamycin (mTOR) signaling pathway, an important negative regulator of autophagy, was demonstrated to be suppressed by DCA treatment. These results may direct the development of novel strategies for the treatment of esophageal squamous carcinoma based on the combined use of DCA and autophagy inhibitors.
Collapse
Affiliation(s)
- Hong-Yu Jia
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| | - He-Nan Wang
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| | - Feng-Yu Xia
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| | - Yan Sun
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| | - Hong-Li Liu
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| | - Li-Li Yan
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| | - Shan-Shan Li
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| | - Dong-Chun Jiang
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| | - Mei-Mei Xu
- Department of Digestive Internal Medicine, First Hospital of Qinhuangdao, Hebei 066000, P.R. China
| |
Collapse
|
40
|
Varshosaz J, Riahi S, Ghassami E, Jahanian-Najafabadi A. Transferrin-targeted poly(butylene adipate)/terephthalate nanoparticles for targeted delivery of 5-fluorouracil in HT29 colorectal cancer cell line. J BIOACT COMPAT POL 2017. [DOI: 10.1177/0883911517690756] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The purpose of this study was to design 5-fluorouracil-loaded poly(butylene adipate)/terephthalate (Ecoflex®) nanoparticles for targeting colorectal cancer. The nanoparticles were prepared by emulsification–solvent evaporation method and optimized by a full factorial design. The effects of polymer and surfactant concentration, surfactant type, and stirrer rate were studied on the particle size, zeta potential, loading efficiency, and release efficiency of nanoparticles. For production of targeted nanoparticles, chitosan was conjugated to transferrin which was then coated on the surface of Ecoflex nanoparticles via electrostatic interactions. The conjugation of transferrin/chitosan was verified by Fourier transform infrared spectroscopy, ultraviolet spectroscopy, and SDS-PAGE (sodium dodecyl sulfate–polyacrylamide gel electrophoresis) methods and quantified by ultraviolet spectroscopy assay. The cytotoxicity of 5-fluorouracil loaded in targeted and non-targeted nanoparticles was studied on human colon adenocarcinoma cell line (HT29), Michigan Cancer Foundation-7 (MCF-7), and human umbilical vein endothelial cells using MTT (thiazolyl blue tetrazolium bromide) assay. The best results were obtained from nanoparticles prepared by 0.2% of the polymer, 2% of Tween 20, and stirrer speed of 17,500 r/min. The successful conjugation of transferrin/chitosan was confirmed by Fourier transform infrared spectrum and SDS-PAGE results and was about 80%. The targeted nanoparticles showed significantly more cytotoxic effects on HT29 cells compared to free 5-fluorouracil and non-targeted nanoparticles. Blocking transferrin receptors resulted in a significantly higher cell survival for targeted nanoparticles which confirmed receptor-mediated cellular uptake of targeted nanoparticles.
Collapse
Affiliation(s)
- Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sara Riahi
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Erfaneh Ghassami
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
41
|
Hosseini M, Kasraian Z, Rezvani HR. Energy metabolism in skin cancers: A therapeutic perspective. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:712-722. [PMID: 28161328 DOI: 10.1016/j.bbabio.2017.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 12/13/2022]
Abstract
Skin cancers are the most common cancers worldwide. The incidence of common skin cancers, including basal cell carcinomas (BCCs), squamous cell carcinomas (SCCs) and melanomas, continues to rise by 5 to 7% per year, mainly due to ultraviolet (UV) exposure and partly because of aging. This suggests an urgent necessity to improve the level of prevention and protection for skin cancers as well as developing new prognostic and diagnostic markers of skin cancers. Moreover, despite innovative therapies especially in the fields of melanoma and carcinomas, new therapeutic options are needed to bypass resistance to targeted therapies or treatment's side effects. Since reprogramming of cellular metabolism is now considered as a hallmark of cancer, some of the recent findings on the role of energy metabolism in skin cancer initiation and progression as well as its effect on the response to targeted therapies are discussed in this review. This article is part of a Special Issue entitled Mitochondria in cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.
Collapse
Affiliation(s)
- Mohsen Hosseini
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Zeinab Kasraian
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Hamid Reza Rezvani
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; Centre de Référence pour les Maladies Rares de la Peau, CHU de Bordeaux, France.
| |
Collapse
|
42
|
Bell H, Parkin E. Pyruvate dehydrogenase kinase inhibition: Reversing the Warburg effect in cancer therapy. INTERNATIONAL JOURNAL OF CANCER THERAPY AND ONCOLOGY 2016. [DOI: 10.14319/ijcto.42.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
43
|
Agnoletto C, Brunelli L, Melloni E, Pastorelli R, Casciano F, Rimondi E, Rigolin GM, Cuneo A, Secchiero P, Zauli G. The anti-leukemic activity of sodium dichloroacetate in p53mutated/null cells is mediated by a p53-independent ILF3/p21 pathway. Oncotarget 2016; 6:2385-96. [PMID: 25544776 PMCID: PMC4385858 DOI: 10.18632/oncotarget.2960] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 12/09/2014] [Indexed: 11/25/2022] Open
Abstract
B-chronic lymphocytic leukemia (B-CLL) patients harboring p53 mutations are invariably refractory to therapies based on purine analogues and have limited treatment options and poor survival. Having recently demonstrated that the mitochondria-targeting small molecule sodium dichloroacetate (DCA) exhibits anti-leukemic activity in p53wild-type B-CLL cells, the aim of this study was to evaluate the effect of DCA in p53mutated B-CLL cells and in p53mutated/null leukemic cell lines. DCA exhibited comparable cytotoxicity in p53wild-type and p53mutated B-CLL patient cell cultures, as well as in p53mutated B leukemic cell lines (MAVER, MEC-1, MEC-2). At the molecular level, DCA promoted the transcriptional induction of p21 in all leukemic cell types investigated, including p53null HL-60. By using a proteomic approach, we demonstrated that DCA up-regulated the ILF3 transcription factor, which is a known regulator of p21 expression. The role of the ILF3/p21 axis in mediating the DCA anti-leukemic activity was underscored by knocking-down experiments. Indeed, transfection with ILF3 and p21 siRNAs significantly decreased both the DCA-induced p21 expression and the DCA-mediated cytotoxicity. Taken together, our results emphasize that DCA is a small molecule that merits further evaluation as a therapeutic agent also for p53mutated leukemic cells, by acting through the induction of a p53-independent pathway.
Collapse
Affiliation(s)
- Chiara Agnoletto
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Laura Brunelli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Elisabetta Melloni
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Roberta Pastorelli
- Institute of Pharmacological Researches, IRCCS "Mario Negri", Milano, Italy
| | - Fabio Casciano
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Erika Rimondi
- Department of Life Science, University of Trieste, Trieste, Italy
| | - Gian Matteo Rigolin
- Department of Medical Sciences, University of Ferrara-Arcispedale S. Anna, Ferrara, Italy
| | - Antonio Cuneo
- Department of Medical Sciences, University of Ferrara-Arcispedale S. Anna, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Giorgio Zauli
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| |
Collapse
|
44
|
Zajac J, Kostrhunova H, Novohradsky V, Vrana O, Raveendran R, Gibson D, Kasparkova J, Brabec V. Potentiation of mitochondrial dysfunction in tumor cells by conjugates of metabolic modulator dichloroacetate with a Pt(IV) derivative of oxaliplatin. J Inorg Biochem 2015; 156:89-97. [PMID: 26780576 DOI: 10.1016/j.jinorgbio.2015.12.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/20/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023]
Abstract
The molecular and cellular mechanisms of enhanced toxic effects in tumor cells of the Pt(IV) derivatives of antitumor oxaliplatin containing axial dichloroacetate (DCA) ligands were investigated. DCA ligands were chosen because DCA has shown great potential as an apoptosis sensitizer and anticancer agent reverting the Wartburg effect. In addition, DCA reverses mitochondrial changes in a wide range of cancers, promoting tumor cell apoptosis in a mitochondrial-dependent pathway. We demonstrate that (i) the transformation of oxaliplatin to its Pt(IV) derivatives containing axial DCA ligands markedly enhances toxicity in cancer cells and helps overcome inherent and acquired resistance to cisplatin and oxaliplatin; (ii) a significant fraction of the intact molecules of DCA conjugates with Pt(IV) derivative of oxaliplatin accumulates in cancer cells where it releases free DCA; (iii) mechanism of biological action of the Pt(IV) derivatives of oxaliplatin containing DCA ligands is connected with the effects of DCA released in cancer cells from the Pt(IV) prodrugs on mitochondria and metabolism of glucose; (iv) treatments with the Pt(IV) derivatives of oxaliplatin containing DCA ligands activate an autophagic response in human colorectal cancer cells; (v) the toxic effects in cancer cells of the Pt(IV) derivatives of oxaliplatin containing DCA ligands can be potentiated if cells are treated with these prodrugs in combination with 5-fluorouracil. These properties of the Pt(IV) derivatives of oxaliplatin containing DCA ligands provide opportunities for further development of new platinum-based agents with the capability of killing cancer cells resistant to conventional antitumor platinum drugs used in the clinic.
Collapse
Affiliation(s)
- Juraj Zajac
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, CZ-61265 Brno, Czech Republic; Department of Biophysics, Faculty of Science, Palacky University, 17. listopadu 12, CZ-77146 Olomouc, Czech Republic
| | - Hana Kostrhunova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - Vojtech Novohradsky
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - Oldrich Vrana
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | - Raji Raveendran
- Institute for Drug Research, School of Pharmacy, The Hebrew University, Jerusalem 91120, Israel
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University, Jerusalem 91120, Israel
| | - Jana Kasparkova
- Department of Biophysics, Faculty of Science, Palacky University, 17. listopadu 12, CZ-77146 Olomouc, Czech Republic
| | - Viktor Brabec
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, CZ-61265 Brno, Czech Republic.
| |
Collapse
|
45
|
Huang CY, Yu LCH. Pathophysiological mechanisms of death resistance in colorectal carcinoma. World J Gastroenterol 2015; 21:11777-11792. [PMID: 26557002 PMCID: PMC4631976 DOI: 10.3748/wjg.v21.i41.11777] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/18/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Colon cancers develop adaptive mechanisms to survive under extreme conditions and display hallmarks of unlimited proliferation and resistance to cell death. The deregulation of cell death is a key factor that contributes to chemoresistance in tumors. In a physiological context, balance between cell proliferation and death, and protection against cell damage are fundamental processes for maintaining gut epithelial homeostasis. The mechanisms underlying anti-death cytoprotection and tumor resistance often bear common pathways, and although distinguishing them would be a challenge, it would also provide an opportunity to develop advanced anti-cancer therapeutics. This review will outline cell death pathways (i.e., apoptosis, necrosis, and necroptosis), and discuss cytoprotective strategies in normal intestinal epithelium and death resistance mechanisms of colon tumor. In colorectal cancers, the intracellular mechanisms of death resistance include the direct alteration of apoptotic and necroptotic machinery and the upstream events modulating death effectors such as tumor suppressor gene inactivation and pro-survival signaling pathways. The autocrine, paracrine and exogenous factors within a tumor microenvironment can also instigate resistance against apoptotic and necroptotic cell death in colon cancers through changes in receptor signaling or transporter uptake. The roles of cyclooxygenase-2/prostaglandin E2, growth factors, glucose, and bacterial lipopolysaccharides in colorectal cancer will be highlighted. Targeting anti-death pathways in the colon cancer tissue might be a promising approach outside of anti-proliferation and anti-angiogenesis strategies for developing novel drugs to treat refractory tumors.
Collapse
|
46
|
Guan DG, Chen HM, Liao SF, Zhao TZ. Combination of temozolomide and Taxol exerts a synergistic inhibitory effect on Taxol‑resistant glioma cells via inhibition of glucose metabolism. Mol Med Rep 2015; 12:7705-11. [PMID: 26459853 DOI: 10.3892/mmr.2015.4405] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/11/2015] [Indexed: 11/06/2022] Open
Abstract
Malignant gliomas, which comprise the most common type of primary malignant brain tumor, are associated with a poor prognosis and quality of life. Paclitaxel (Taxol) and temozolomide (TMZ) are Food and Drug Administration‑approved anticancer agents, which are known to have therapeutic applications in various malignancies. However, similar to other chemotherapeutic agents, the development of resistance to TMZ and Taxol is common. The aim of the present study was to investigate the regulation of glucose metabolism by TMZ and Taxol in glioma cells. The results demonstrated that glioma cells exhibit decreased glucose uptake and lactate production in response to treatment with TMZ; however, glucose metabolism was increased in response to Taxol treatment. Following analysis of TMZ‑ and Taxol‑resistant cell lines, it was reported that glucose metabolism was decreased in the TMZ‑resistant cells, but was increased in the Taxol‑resistant cells. Notably, a combination of TMZ and Taxol exerted synergistic inhibitory effects on Taxol‑resistant glioma cells. However, the synergistic phenotype was not observed following treatment with a combination of 5‑fluorouracil and Taxol. Furthermore, restoration of glucose metabolism by overexpression of glucose transporter 1 in Taxol‑resistant cells resulted in regained resistance to Taxol. Therefore, the present study proposes a novel mechanism accounting for the synergistic effects of Taxol and TMZ co‑treatment, which may contribute to the development of therapeutic strategies for overcoming chemoresistance in patients with cancer.
Collapse
Affiliation(s)
- Ding-Guo Guan
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Han-Min Chen
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Sheng-Fang Liao
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Tian-Zhi Zhao
- Department of Neurosurgery, Affiliated Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shanxi 710038, P.R. China
| |
Collapse
|
47
|
Shi DB, Li XX, Zheng HT, Li DW, Cai GX, Peng JJ, Gu WL, Guan ZQ, Xu Y, Cai SJ. Icariin-mediated inhibition of NF-κB activity enhances the in vitro and in vivo antitumour effect of 5-fluorouracil in colorectal cancer. Cell Biochem Biophys 2015; 69:523-30. [PMID: 24435883 DOI: 10.1007/s12013-014-9827-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Colorectal cancer (CRC) is an aggressive malignancy that has a poor prognosis. 5-Fluorouracil (5-FU) is a first line chemotherapeutic medication used in the treatment of gallbladder cancer; however, the efficacy is below satisfactory. Icariin is a natural compound that is conventionally reported to have activity against a variety of cancers. This study was carried out to investigate the anti-cancer effect of icariin in CRC cells and to determine whether the compound can enhance the antitumour activity of 5-FU. Cell proliferation and apoptosis were measured using an MTT assay and flow cytometry, respectively. The activity of transcription factor NF-κB was determined by EMSA method. The expression of apoptosis- and proliferation-related proteins was determined by western blotting. The in vivo antitumour effect of combination treatment with icariin and 5-FU on CRC was also assessed using a murine model of CRC. Icariin sensitized the CRC cells to 5-FU both in vitro and in vivo. The antitumour activity of icariin and its potentiating effect on the antitumour activity of 5-FU implicated the suppression of NF-κB activity and consequent down-regulation of the gene products regulated by NF-κB. Our results showed that icariin, suppressed tumour growth and enhanced the antitumour activity of 5-FU in CRC by inhibiting NF-κB activity. Therefore, we suggest that combination of icariin with 5-FU might offer a therapeutic benefit to the patients with CRC; however, further studies are required to ascertain this proposition.
Collapse
Affiliation(s)
- De-Bing Shi
- Department of Colorectal Surgery, Cancer Hospital, Fudan University, Shanghai, 200032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
A Novel Pharmacological Method to Study the Chinese Medicinal Formula Hua-Zheng-Hui-Sheng-Dan. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:436807. [PMID: 26421050 PMCID: PMC4569805 DOI: 10.1155/2015/436807] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 08/10/2015] [Accepted: 08/16/2015] [Indexed: 02/05/2023]
Abstract
Objectives. Hua-Zheng-Hui-Sheng-Dan (HZHSD) was used as an experimental model to explore research methods of large formulae in traditional Chinese medicine (TCM) using current molecular biology approaches. Materials and Methods. The trypan blue exclusion assay was used to determine cell viability and cell numbers. Flow cytometry was used to assess cell cycle distribution and apoptosis. The concentration of cyclin D1 was analyzed by enzyme-linked immunosorbent assay. The median effect principle was used in drug combination studies. An orthogonal experimental design was used to estimate the effects of each herb at different concentrations. The HeLa xenograft mouse model was used to compare the antitumor activity of drugs in vivo. Results. Among the 35 herbs that comprise HZHSD, Radix Rehmanniae Preparata (RRP), Caesalpinia sappan (CS), Evodia rutaecarpa (ER), Folium Artemisiae Argyi (FAA), Leonurus japonicus Houtt (LJH), Tumeric (Tu), Radix Paeoniae Alba (RPA), and Trogopterus Dung (TD) effectively inhibited the proliferation of HeLa and SKOV3 cells. Only RRR had an effect on HeLa and SKOV3 cell viability. According to the median effect principle, Angelica sinensis (Oliv.) (AS), Tabanus (Ta), and Pollen Typhae (PT), which were proven to have a significant synergistic inhibitory effect on the proliferation of HeLa cells, were added to the original eight positive herbs. The combination of RPA and AS had a synergistic effect on inducing cell cycle S phase arrest and decreasing intracellular cyclin D1 in HeLa cells. By orthogonal experimental design, LJH and Tu were considered unnecessary herbs. The small formula (SHZHSD) consisted of RPA, AS, RRR, Ta., TD, PT, ER, CS, and FAA and was able to inhibit cell proliferation and induce cell apoptosis. The antitumor effects of HZHSD and SHZHSD were also compared in vivo. Conclusions. Through molecular biology approaches both in vitro and in vivo, research into single drugs, and analysis using the median effect principle and orthogonal experimental design, the small formula (SHZHSD) was determined from the original formula (HZHSD). SHZHSD exhibited superior antitumor activity compared with the original formula both in vitro and in vivo.
Collapse
|
49
|
Lee M, Yoon JH. Metabolic interplay between glycolysis and mitochondrial oxidation: The reverse Warburg effect and its therapeutic implication. World J Biol Chem 2015; 6:148-61. [PMID: 26322173 PMCID: PMC4549759 DOI: 10.4331/wjbc.v6.i3.148] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 05/26/2015] [Accepted: 07/21/2015] [Indexed: 02/05/2023] Open
Abstract
Aerobic glycolysis, i.e., the Warburg effect, may contribute to the aggressive phenotype of hepatocellular carcinoma. However, increasing evidence highlights the limitations of the Warburg effect, such as high mitochondrial respiration and low glycolysis rates in cancer cells. To explain such contradictory phenomena with regard to the Warburg effect, a metabolic interplay between glycolytic and oxidative cells was proposed, i.e., the "reverse Warburg effect". Aerobic glycolysis may also occur in the stromal compartment that surrounds the tumor; thus, the stromal cells feed the cancer cells with lactate and this interaction prevents the creation of an acidic condition in the tumor microenvironment. This concept provides great heterogeneity in tumors, which makes the disease difficult to cure using a single agent. Understanding metabolic flexibility by lactate shuttles offers new perspectives to develop treatments that target the hypoxic tumor microenvironment and overcome the limitations of glycolytic inhibitors.
Collapse
|
50
|
Abstract
Dichloroacetate (DCA) is a metabolic reprogramming agent that reverses the Warburg effect, causing cancer cells to couple glycolysis to oxidative phosphorylation. This has been shown to induce apoptosis and reduce the growth of various types of cancer but not normal cells. Colorectal cancer cells HCT116, HCT116 p53(-/-), and HCT116 Bax(-/-), were treated with DCA in vitro. Response to treatment was determined by measuring PDH phosphorylation, apoptosis, proliferation, and cell cycle. Molecular changes associated with these responses were determined using western immunoblotting and quantitative PCR. Treatment with 20 mM DCA did not increase apoptosis, despite decreasing levels of anti-apoptotic protein Mcl-1 after 6 h, in any of the cell lines observed. Mcl-1 expression was stabilized with MG-132, an inhibitor of proteasomal degradation. A decrease in Mcl-1 correlated with a decrease in proliferation, both of which showed dose-dependence in DCA treated cells. Cells showed nuclear localization of Mcl-1, however cell cycle was unaffected by DCA treatment. These data suggest that a reduction in the prosurvival Bcl-2 family member Mcl-1 due to increased proteasomal degradation is correlated with the ability of DCA to reduce proliferation of HCT116 human colorectal cancer cells without causing apoptosis.
Collapse
|