1
|
Nagargoje AA, Deshmukh TR, Shaikh MH, Khedkar VM, Shingate BB. Anticancer perspectives of monocarbonyl analogs of curcumin: A decade (2014-2024) review. Arch Pharm (Weinheim) 2024; 357:e2400197. [PMID: 38895952 DOI: 10.1002/ardp.202400197] [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: 03/16/2024] [Revised: 05/13/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024]
Abstract
Monocarbonyl analogs of curcumin (MACs) represent structurally modified versions of curcumin. The existing literature indicates that MACs exhibit enhanced anticancer properties compared with curcumin. Numerous research articles in recent years have emphasized the significance of MACs as effective anticancer agents. This review focuses on the latest advances in the anticancer potential of MACs, from 2014 to 2024, including discussions on their mechanism of action, structure-activity relationship (SAR), and in silico molecular docking studies.
Collapse
Affiliation(s)
- Amol A Nagargoje
- Department of Chemistry, Dr. Babasaheb Ambedkar Marathwada University, Chhatrapati Sambhajinagar (Aurangabad), Maharashtra, India
- Department of Chemistry, Khopoli Municipal Council College, Khopoli, Maharashtra, India
| | - Tejshri R Deshmukh
- Department of Chemistry, Dr. Babasaheb Ambedkar Marathwada University, Chhatrapati Sambhajinagar (Aurangabad), Maharashtra, India
| | - Mubarak H Shaikh
- Department of Chemistry, Dr. Babasaheb Ambedkar Marathwada University, Chhatrapati Sambhajinagar (Aurangabad), Maharashtra, India
- Department of Chemistry, Radhabai Kale Mahila Mahavidyalaya, Ahmednagar, Maharashtra, India
| | - Vijay M Khedkar
- School of Pharmacy, Vishwakarma University, Pune, Maharashtra, India
| | - Bapurao B Shingate
- Department of Chemistry, Dr. Babasaheb Ambedkar Marathwada University, Chhatrapati Sambhajinagar (Aurangabad), Maharashtra, India
| |
Collapse
|
2
|
Zheng X, Tang P, Li H, Ye T, Zhu X, He W, Cheng L, Cheng H. Cucurbitacin E elicits apoptosis in laryngeal squamous cell carcinoma by enhancing reactive oxygen species-regulated mitochondrial dysfunction and endoplasmic reticulum stress. Am J Cancer Res 2024; 14:3905-3921. [PMID: 39267666 PMCID: PMC11387858 DOI: 10.62347/hpqq9412] [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: 06/04/2024] [Accepted: 08/18/2024] [Indexed: 09/15/2024] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a prevalent head and neck neoplasm with escalating global morbidity and mortality rates. Despite the increasing burden of LSCC, the drugs currently approved for its treatment are limited. Therefore, it is necessary to identify novel and promising drugs that target LSCC. Cucurbitacin E (CuE) is a naturally oxygenated tetracyclic triterpenoid that suppresses several cancers. However, its anti-LSCC activity and the molecular mechanisms of action remain unclear. This study explored its impact on LSCC, revealing cell viability attenuation and apoptosis enhancement in vitro. Further investigations indicated that CuE significantly decreased mitochondrial membrane potential, thereby promoting cytochrome c release, increasing cleaved-Caspase 3 and cleaved-PARP levels, and triggering mitochondria-dependent apoptosis. Concurrently, exposure of LSCC cells to CuE enhanced endoplasmic reticulum (ER) stress, mobilized the protein kinase RNA-like endoplasmic reticulum kinase/initiation factor 2a/ATF4/C-EBP homologous protein pathway, and induced LSCC cell apoptosis. Finally, CuE markedly elevated intracellular reactive oxygen species (ROS) levels. When ROS were eliminated with N-acetylcysteine, CuE-mediated mitochondrial dysfunction, ER stress, and cell apoptosis were nearly abolished. Similar outcomes were observed in murine LSCC models. Together, these results highlight that CuE suppresses proliferation while triggering apoptosis in LSCC cells via ROS-regulated mitochondrial dysfunction and the ER stress pathway. Hence, CuE may serve as a promising candidate for LCSS treatment.
Collapse
Affiliation(s)
- Xucai Zheng
- Department of Oncology, The Second Hospital of Anhui Medical University Hefei 230601, Anhui, China
- Department of Head, Neck and Breast Surgery, The First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital Hefei 230031, Anhui, China
| | - Puze Tang
- Bachelor of Science in Mathematics, University of Liverpool United Kingdom
| | - Hui Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Tingbo Ye
- Department of Head, Neck and Breast Surgery, The First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital Hefei 230031, Anhui, China
| | - Xu Zhu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Wei He
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Ling Cheng
- Medical Intensive Care Unit, The First Affiliated Hospital of Anhui University of Chinese Medicine Hefei 230031, Anhui, China
| | - Huaidong Cheng
- Department of Oncology, The Second Hospital of Anhui Medical University Hefei 230601, Anhui, China
- Department of Oncology, Shenzhen Hospital of Southern Medical University Shenzhen 518000, Guangdong, China
| |
Collapse
|
3
|
Cao Y, Zhou X, Nie Q, Zhang J. Inhibition of the thioredoxin system for radiosensitization therapy of cancer. Eur J Med Chem 2024; 268:116218. [PMID: 38387331 DOI: 10.1016/j.ejmech.2024.116218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024]
Abstract
Radiotherapy (RT) stands as a cornerstone in the clinical armamentarium against various cancers due to its proven efficacy. However, the intrinsic radiation resistance exhibited by cancer cells, coupled with the adverse effects of RT on normal tissues, often compromises its therapeutic potential and leads to unwanted side effects. This comprehensive review aims to consolidate our understanding of how radiosensitizers inhibit the thioredoxin (Trx) system in cellular contexts. Notable radiosensitizers, including gold nanoparticles (GNPs), gold triethylphosphine cyanide ([Au(SCN) (PEt3)]), auranofin, ceria nanoparticles (CONPs), curcumin and its derivatives, piperlongamide, indolequinone derivatives, micheliolide, motexafin gadolinium, and ethane selenide selenidazole derivatives (SeDs), are meticulously elucidated in terms of their applications in radiotherapy. In this review, the sensitization mechanisms and the current research progress of these radiosensitizers are discussed in detail, with the overall aim of providing valuable insights for the judicious application of Trx system inhibitors in the field of cancer radiosensitization therapy.
Collapse
Affiliation(s)
- Yisheng Cao
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Xiedong Zhou
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Qiuying Nie
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
4
|
Gao Y, Gong Y, Lu J, Hao H, Shi X. Targeting YAP1 to improve the efficacy of immune checkpoint inhibitors in liver cancer: mechanism and strategy. Front Immunol 2024; 15:1377722. [PMID: 38550587 PMCID: PMC10972981 DOI: 10.3389/fimmu.2024.1377722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
Liver cancer is the third leading of tumor death, including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). Immune checkpoint inhibitors (ICIs) are yielding much for sufferers to hope for patients, but only some patients with advanced liver tumor respond. Recent research showed that tumor microenvironment (TME) is critical for the effectiveness of ICIs in advanced liver tumor. Meanwhile, metabolic reprogramming of liver tumor leads to immunosuppression in TME. These suggest that regulating the abnormal metabolism of liver tumor cells and firing up TME to turn "cold tumor" into "hot tumor" are potential strategies to improve the therapeutic effect of ICIs in liver tumor. Previous studies have found that YAP1 is a potential target to improve the efficacy of anti-PD-1 in HCC. Here, we review that YAP1 promotes immunosuppression of TME, mainly due to the overstimulation of cytokines in TME by YAP1. Subsequently, we studied the effects of YAP1 on metabolic reprogramming in liver tumor cells, including glycolysis, gluconeogenesis, lipid metabolism, arachidonic acid metabolism, and amino acid metabolism. Lastly, we summarized the existing drugs targeting YAP1 in the treatment of liver tumor, including some medicines from natural sources, which have the potential to improve the efficacy of ICIs in the treatment of liver tumor. This review contributed to the application of targeted YAP1 for combined therapy with ICIs in liver tumor patients.
Collapse
Affiliation(s)
- Yuting Gao
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yi Gong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Junlan Lu
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Huiqin Hao
- Chinese Medicine Gene Expression Regulation Laboratory, State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
- Basic Laboratory of Integrated Traditional Chinese and Western, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xinli Shi
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| |
Collapse
|
5
|
Besasie BD, Saha A, DiGiovanni J, Liss MA. Effects of curcumin and ursolic acid in prostate cancer: A systematic review. Urologia 2024; 91:90-106. [PMID: 37776274 DOI: 10.1177/03915603231202304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
The major barriers to phytonutrients in prostate cancer therapy are non-specific mechanisms and bioavailability issues. Studies have pointed to a synergistic combination of curcumin (CURC) and ursolic acid (UA). We investigate this combination using a systematic review process to assess the most likely mechanistic pathway and human testing in prostate cancer. We used the PRISMA statement to screen titles, abstracts, and the full texts of relevant articles and performed a descriptive analysis of the literature reviewed for study inclusion and consensus of the manuscript. The most common molecular and cellular pathway from articles reporting on the pathways and effects of CURC (n = 173) in prostate cancer was NF-κB (n = 25, 14.5%). The most common molecular and cellular pathway from articles reporting on the pathways and effects of UA (n = 24) in prostate cancer was caspase 3/caspase 9 (n = 10, 41.6%). The three most common molecular and cellular pathway from articles reporting on the pathways and effects of both CURC and UA (n = 193) in prostate cancer was NF-κB (n = 28, 14.2%), Akt (n = 22, 11.2%), and androgen (n = 19, 9.6%). Therefore, we have identified the potential synergistic target pathways of curcumin and ursolic acid to involve NF-κB, Akt, androgen receptors, and apoptosis pathways. Our review highlights the limited human studies and specific effects in prostate cancer.
Collapse
Affiliation(s)
- Benjamin D Besasie
- Department of Urology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Achinto Saha
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, USA
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, USA
| | - Michael A Liss
- Department of Urology, University of Texas Health San Antonio, San Antonio, TX, USA
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, USA
- Department of Urology, South Texas Veterans Healthcare System, USA
| |
Collapse
|
6
|
Gaur T, Ali A, Sharma D, Gupta SK, Gota V, Bagal B, Platzbeckar U, Mishra R, Dutt A, Khattry N, Mills K, Hassan MI, Sandur S, Hasan SK. Mitocurcumin utilizes oxidative stress to upregulate JNK/p38 signaling and overcomes Cytarabine resistance in acute myeloid leukemia. Cell Signal 2024; 114:111004. [PMID: 38048856 DOI: 10.1016/j.cellsig.2023.111004] [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: 08/18/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023]
Abstract
Acute myeloid leukemia (AML) is a type of blood cancer that is characterized by the rapid growth of abnormal myeloid cells. The goal of AML treatment is to eliminate the leukemic blasts, which is accomplished through intensive chemotherapy. Cytarabine is a key component of the standard induction chemotherapy regimen for AML. However, despite a high remission rate, 70-80% of AML patients relapse and develop resistance to Cytarabine, leading to poor clinical outcomes. Mitocurcumin (MitoC), a derivative of curcumin that enters mitochondria, leading to a drop in mitochondrial membrane potential and mitophagy induction. Further, it activates oxidative stress-mediated JNK/p38 signaling to induce apoptosis. MitoC demonstrated a preferential ability to kill leukemic cells from AML cell lines and patient-derived leukemic blasts. RNA sequencing data suggests perturbation of DNA damage response and cell proliferation pathways in MitoC-treated AML. Elevated reactive oxygen species (ROS) in MitoC-treated AML cells resulted in significant DNA damage and cell cycle arrest. Further, MitoC treatment resulted in ROS-mediated enhanced levels of p21, which leads to suppression of CHK1, RAD51, Cyclin-D and c-Myc oncoproteins, potentially contributing to Cytarabine resistance. Combinatorial treatment of MitoC and Cytarabine has shown synergism, increased apoptosis, and enhanced DNA damage. Using AML xenografts, a significant reduction of hCD45+ cells was observed in AML mice bone marrow treated with MitoC (mean 0.6%; range0.04%-3.56%) compared to control (mean 38.2%; range10.1%-78%), p = 0.03. The data suggest that MitoC exploits stress-induced leukemic oxidative environment to up-regulate JNK/p38 signaling to lead to apoptosis and can potentially overcome Cytarabine resistance via ROS/p21/CHK1 axis.
Collapse
Affiliation(s)
- Tarang Gaur
- Hasan Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
| | - Ahlam Ali
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Deepak Sharma
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India; Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Saurabh Kumar Gupta
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India; Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, Maharashtra, India
| | - Vikram Gota
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India; Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, Maharashtra, India
| | - Bhausaheb Bagal
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India; Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400014, India
| | - Uwe Platzbeckar
- Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital Leipzig, Johannisallee 32, D-04103 Leipzig, Germany
| | - Rohit Mishra
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India; Dutt Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
| | - Amit Dutt
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India; Dutt Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
| | - Navin Khattry
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India; Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400014, India
| | - Ken Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Santosh Sandur
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India; Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Syed K Hasan
- Hasan Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
7
|
Zhao Y, Ye X, Xiong Z, Ihsan A, Ares I, Martínez M, Lopez-Torres B, Martínez-Larrañaga MR, Anadón A, Wang X, Martínez MA. Cancer Metabolism: The Role of ROS in DNA Damage and Induction of Apoptosis in Cancer Cells. Metabolites 2023; 13:796. [PMID: 37512503 PMCID: PMC10383295 DOI: 10.3390/metabo13070796] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer is a huge challenge for people worldwide. High reactive oxygen species (ROS) levels are a recognized hallmark of cancer and an important aspect of cancer treatment research. Abnormally elevated ROS levels are often attributable to alterations in cellular metabolic activities and increased oxidative stress, which affects both the development and maintenance of cancer. Moderately high levels of ROS are beneficial to maintain tumor cell genesis and development, while toxic levels of ROS have been shown to be an important force in destroying cancer cells. ROS has become an important anticancer target based on the proapoptotic effect of toxic levels of ROS. Therefore, this review summarizes the role of increased ROS in DNA damage and the apoptosis of cancer cells caused by changes in cancer cell metabolism, as well as various anticancer therapies targeting ROS generation, in order to provide references for cancer therapies based on ROS generation.
Collapse
Affiliation(s)
- Yongxia Zhao
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaochun Ye
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhifeng Xiong
- Department of Animal Nutrition and Feed Science, Huazhong Agricultural University, Wuhan 430070, China
| | - Awais Ihsan
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Sahiwal 57000, Pakistan
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Marta Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| | - María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid (UCM), and Research Institute Hospital 12 de Octubre (i+12), 28040 Madrid, Spain
| |
Collapse
|
8
|
Nizami ZN, Aburawi HE, Semlali A, Muhammad K, Iratni R. Oxidative Stress Inducers in Cancer Therapy: Preclinical and Clinical Evidence. Antioxidants (Basel) 2023; 12:1159. [PMID: 37371889 DOI: 10.3390/antiox12061159] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Reactive oxygen species (ROS) are metabolic byproducts that regulate various cellular processes. However, at high levels, ROS induce oxidative stress, which in turn can trigger cell death. Cancer cells alter the redox homeostasis to facilitate protumorigenic processes; however, this leaves them vulnerable to further increases in ROS levels. This paradox has been exploited as a cancer therapeutic strategy with the use of pro-oxidative drugs. Many chemotherapeutic drugs presently in clinical use, such as cisplatin and doxorubicin, induce ROS as one of their mechanisms of action. Further, various drugs, including phytochemicals and small molecules, that are presently being investigated in preclinical and clinical studies attribute their anticancer activity to ROS induction. Consistently, this review aims to highlight selected pro-oxidative drugs whose anticancer potential has been characterized with specific focus on phytochemicals, mechanisms of ROS induction, and anticancer effects downstream of ROS induction.
Collapse
Affiliation(s)
- Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Hanan E Aburawi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire-Université Laval, Quebec, QC G1V 0A6, Canada
| | - Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| |
Collapse
|
9
|
Zhu H, Gao M, Sun W, Liu H, Xu S, Li X. ROS/ER stress contributes to trimethyltin chloride-mediated hepatotoxicity; Tea polyphenols alleviate apoptosis and immunosuppression. Comp Biochem Physiol C Toxicol Pharmacol 2023; 263:109505. [PMID: 36370998 DOI: 10.1016/j.cbpc.2022.109505] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022]
Abstract
Trimethyltin chloride (TMT) is an organotin-based contaminant present in the water environment that poses a great threat to aquatic organisms and humans. The liver is the detoxification organ of the body and TMT exposure accumulates in the liver. Tea polyphenol (TP) is a natural antioxidant extracted from tea leaves and has been widely used as a food and feed additive. To investigate the mechanism of toxicity caused by TMT exposure on grass carp hepatocytes (L8824 cells) and the mitigating effect of TP, we established a hepatocyte model of TMT toxicity and/or TP treatment. L8824 cells were treated with 0.5 μM of TMT and/or 4 μg/mL of TP for 24 h and assayed for relevant indices. The results showed that TMT exposure caused oxidative stress, resulting in increased intracellular ROS content, resulting in intracellular ROS accumulation and increased MDA content, and inhibiting the activities of T-AOC, SOD, CAT, and GSH. Meanwhile, TMT exposure activated the endoplasmic reticulum apoptotic signaling pathway, resulting in abnormal expression of GRP78, ATF-6, IRE1, PERK, Caspase-3 and Caspase-12. In addition, TMT exposure also led to up-regulation of cytokines IL-1β, IL-6, TNF-α, and decreased expression of IL-2, IFN-γ, and antimicrobial peptides Hepcidin, β-defensin, and LEAP2. However, the addition of TP could mitigate the above changes. In conclusion, TP can alleviate TMT exposure-mediated hepatotoxicity by inhibiting ROS/ER stress in L8824 cells. In addition, this trial enriches the cytotoxicity study of TMT and provides a new theoretical basis for the use of TP as a mitigating agent for TMT.
Collapse
Affiliation(s)
- Huijun Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Meichen Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Wenying Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Huanyi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Xiaojing Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
10
|
Pantiora P, Furlan V, Matiadis D, Mavroidi B, Perperopoulou F, Papageorgiou AC, Sagnou M, Bren U, Pelecanou M, Labrou NE. Monocarbonyl Curcumin Analogues as Potent Inhibitors against Human Glutathione Transferase P1-1. Antioxidants (Basel) 2022; 12:antiox12010063. [PMID: 36670925 PMCID: PMC9854774 DOI: 10.3390/antiox12010063] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
The isoenzyme of human glutathione transferase P1-1 (hGSTP1-1) is involved in multi-drug resistance (MDR) mechanisms in numerous cancer cell lines. In the present study, the inhibition potency of two curcuminoids and eleven monocarbonyl curcumin analogues against hGSTP1-1 was investigated. Demethoxycurcumin (Curcumin II) and three of the monocarbonyl curcumin analogues exhibited the highest inhibitory activity towards hGSTP1-1 with IC50 values ranging between 5.45 ± 1.08 and 37.72 ± 1.02 μM. Kinetic inhibition studies of the most potent inhibitors demonstrated that they function as non-competitive/mixed-type inhibitors. These compounds were also evaluated for their toxicity against the prostate cancer cells DU-145. Interestingly, the strongest hGSTP1-1 inhibitor, (DM96), exhibited the highest cytotoxicity with an IC50 of 8.60 ± 1.07 μΜ, while the IC50 values of the rest of the compounds ranged between 44.59-48.52 μΜ. Structural analysis employing molecular docking, molecular dynamics (MD) simulations, and binding-free-energy calculations was performed to study the four most potent curcumin analogues as hGSTP1-1 inhibitors. According to the obtained computational results, DM96 exhibited the lowest binding free energy, which is in agreement with the experimental data. All studied curcumin analogues were found to form hydrophobic interactions with the residue Gln52, as well as hydrogen bonds with the nearby residues Gln65 and Asn67. Additional hydrophobic interactions with the residues Phe9 and Val36 as well as π-π stacking interaction with Phe9 contributed to the superior inhibitory activity of DM96. The van der Waals component through shape complementarity was found to play the most important role in DM96-inhibitory activity. Overall, our results revealed that the monocarbonyl curcumin derivative DM96 acts as a strong hGSTP1-1 inhibitor, exerts high prostate cancer cell cytotoxicity, and may, therefore, be exploited for the suppression and chemosensitization of cancer cells. This study provides new insights into the development of safe and effective GST-targeted cancer chemosensitizers.
Collapse
Affiliation(s)
- Panagiota Pantiora
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, GR-11855 Athens, Greece
- Institute of Biosciences & Applications, NCSR “Demokritos”, 15310 Athens, Greece
| | - Veronika Furlan
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia
| | - Dimitris Matiadis
- Institute of Biosciences & Applications, NCSR “Demokritos”, 15310 Athens, Greece
| | - Barbara Mavroidi
- Institute of Biosciences & Applications, NCSR “Demokritos”, 15310 Athens, Greece
| | - Fereniki Perperopoulou
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, GR-11855 Athens, Greece
| | | | - Marina Sagnou
- Institute of Biosciences & Applications, NCSR “Demokritos”, 15310 Athens, Greece
| | - Urban Bren
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000 Koper, Slovenia
- Institute of Environmental Protection and Sensors, Beloruska Ulica 7, SI-2000 Maribor, Slovenia
| | - Maria Pelecanou
- Institute of Biosciences & Applications, NCSR “Demokritos”, 15310 Athens, Greece
| | - Nikolaos E. Labrou
- Laboratory of Enzyme Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos Street, GR-11855 Athens, Greece
- Correspondence: ; Tel./Fax: +30-2105294208
| |
Collapse
|
11
|
Boddu A, Obireddy SR, Zhang D, Rao KSVK, Lai WF. ROS-generating, pH-responsive and highly tunable reduced graphene oxide-embedded microbeads showing intrinsic anticancer properties and multi-drug co-delivery capacity for combination cancer therapy. Drug Deliv 2022; 29:2481-2490. [PMID: 35912830 PMCID: PMC9347472 DOI: 10.1080/10717544.2022.2100512] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The development of effective carriers enabling combination cancer therapy is of practical importance due to its potential to enhance the effectiveness of cancer treatment. However, most of the reported carriers are monofunctional in nature. The carriers that can be applied to concomitantly mediate multiple treatment modalities are highly deficient. This study fills this gap by reporting the design and fabrication of ROS-generating carbohydrate-based pH-responsive beads with intrinsic anticancer therapy and multidrug co-delivery capacity for combination cancer therapy. Sodium alginate (SA) microspheres and reduced graphene oxide (rGO)-embedded chitosan (CS) beads are developed via emulsion-templated ionic gelation for a combination therapy involving co-delivery of curcumin (CUR) and 5-fluororacil (5-FU). Drug-encapsulated microbeads are characterized by FTIR, DSC, TGA, XRD, and SEM. 5-FU and CUR-encapsulated microbeads are subjected to in vitro drug release studies at pH 6.8 and 1.2 at 37 °C. Various release kinetic parameters are evaluated. The results show that the Korsmeyer-Peppas model and non-Fickian release kinetics are best suited. The microspheres and microbeads are found to effectively act against MCF7 cells and show intrinsic anticancer capacity. These results indicate the promising performance of our beads in mediating combination drug therapy to improve the effectiveness of cancer treatment.
Collapse
Affiliation(s)
- Adilakshmi Boddu
- Polymer Biomaterial Design and Synthesis Laboratory, Department of Chemistry, Yogi Vemana University, Kadapa, India
| | - Sreekanth Reddy Obireddy
- Department of Chemistry, Sri Krishnadeveraya University, Anantapuramu, India.,Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang, China
| | - Dahong Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang, China
| | - K S V Krishna Rao
- Polymer Biomaterial Design and Synthesis Laboratory, Department of Chemistry, Yogi Vemana University, Kadapa, India
| | - Wing-Fu Lai
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang, China.,Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
12
|
Shiau JP, Chuang YT, Tang JY, Yang KH, Chang FR, Hou MF, Yen CY, Chang HW. The Impact of Oxidative Stress and AKT Pathway on Cancer Cell Functions and Its Application to Natural Products. Antioxidants (Basel) 2022; 11:1845. [PMID: 36139919 PMCID: PMC9495789 DOI: 10.3390/antiox11091845] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023] Open
Abstract
Oxidative stress and AKT serine-threonine kinase (AKT) are responsible for regulating several cell functions of cancer cells. Several natural products modulate both oxidative stress and AKT for anticancer effects. However, the impact of natural product-modulating oxidative stress and AKT on cell functions lacks systemic understanding. Notably, the contribution of regulating cell functions by AKT downstream effectors is not yet well integrated. This review explores the role of oxidative stress and AKT pathway (AKT/AKT effectors) on ten cell functions, including apoptosis, autophagy, endoplasmic reticulum stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, senescence, migration, and cell-cycle progression. The impact of oxidative stress and AKT are connected to these cell functions through cell function mediators. Moreover, the AKT effectors related to cell functions are integrated. Based on this rationale, natural products with the modulating abilities for oxidative stress and AKT pathway exhibit the potential to regulate these cell functions, but some were rarely reported, particularly for AKT effectors. This review sheds light on understanding the roles of oxidative stress and AKT pathway in regulating cell functions, providing future directions for natural products in cancer treatment.
Collapse
Affiliation(s)
- Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
| | - Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
13
|
Nelson VK, Pullaiah CP, Saleem Ts M, Roychoudhury S, Chinnappan S, Vishnusai B, Ram Mani R, Birudala G, Bottu KS. Natural Products as the Modulators of Oxidative Stress: An Herbal Approach in the Management of Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:161-179. [PMID: 36472822 DOI: 10.1007/978-3-031-12966-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer is the most commonly diagnosed and frequently occurred cancer in the males globally. The current treatment strategies available to treat prostate cancer are not much effective and express various adverse effects. Hence, there is an urgent need to identify novel treatment that can improve patient outcome. From times immemorial, natural products are highly recognized for novel drug development for various diseases including cancer. Cancer cells generally maintain higher basal levels of reactive oxygen species (ROS) when compared to normal cells due to its high metabolic rate. However, initiation of excess intracellular ROS production can not be tolerated by the cancer cells and induce several cell death signals which are in contrast to normal cells. Therefore, small molecules of natural origin that induce ROS can potentially kill cancer cells in specific and provide a better opportunity to develop a novel drug therapy. In this review, we elaborated various classes of medicinal compounds and their mechanism of killing prostate cancer cells through direct or indirect ROS generation. This can generate a novel thought to develop promising drug candidate to treat prostate cancer patients.
Collapse
Affiliation(s)
- Vinod K Nelson
- Department of Pharmaceutical Chemistry, Raghavendra Institute of Pharmaceutical Education and Research (Autonomous), Anantapuramu, Andhra Pradesh, India.
| | - Chitikela P Pullaiah
- Department of Pharmacology, Siddha Central Research Institute, Central Council for Research in Siddha, Ministry of AYUSH, Chennai, Tamil Nadu, India
| | - Mohammed Saleem Ts
- College of Pharmacy, Riyadh ELM University, Riyadh, Kingdom of Saudi Arabia, Riyadh
| | | | - Sasikala Chinnappan
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur, Malaysia
| | - Beere Vishnusai
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Ravishankar Ram Mani
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur, Malaysia
| | - Geetha Birudala
- Faculty of Pharmacy, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Kavya Sree Bottu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| |
Collapse
|
14
|
Mohsin A, Haneef K, Ilyas A, Zarina S, Hashim Z. Oxidative Stress Induced Cell Cycle Arrest: Potential Role of PRX-2 and GSTP-1 as Therapeutic Targets in Hepatocellular Carcinoma. Protein Pept Lett 2021; 28:1323-1329. [PMID: 34749598 DOI: 10.2174/0929866528666211105105953] [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: 08/05/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The increasing incidence and mortality rate of HCC is a major concern, especially for developing countries of the world. Hence, extensive research is being carried out in order to explore new approaches for developing successful therapeutic strategies for HCC. The controversial role of oxidative stress in the prognosis and treatment of various diseases such as cancer has become the area of great interest and intrigue for many scientists throughout the world. OBJECTIVE We aim to investigate the role of induced oxidative stress on the suppression of HCC Huh-7 cancerous cells as therapeutic approach. METHODS Induction of oxidative stress via H2O2 treatment produced cell cytotoxicity in a dose dependent manner and also led to the over expression of GSTP-1 and PRX-2. The expression of GSTP-1 and PRX-2 was compared in HCC Huh-7 treated, untreated cells and normal hepatocytes using immunocytochemistry. Furthermore, the effects of oxidative stress on cell cycle arrest were also studied through flow cytometry. RESULTS Our study demonstrated the inhibition of cancer cell proliferation as a result of H2O2 induction by arresting the cell cycle at G2 phase. CONCLUSION The induction of oxidative stress could be a potential therapeutic approach for treating HCC in the future. GSTP-1 and PRX-2 can serve as substantial therapeutic targets for the treatment of HCC.
Collapse
Affiliation(s)
- Abeer Mohsin
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| | - Kanwal Haneef
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| | - Amber Ilyas
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| | - Shamshad Zarina
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| | - Zehra Hashim
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| |
Collapse
|
15
|
Abd Wahab NA, Abas F, Othman I, Naidu R. Diarylpentanoid (1,5-bis(4-hydroxy-3-methoxyphenyl)-1,4-pentadiene-3-one) (MS13) Exhibits Anti-proliferative, Apoptosis Induction and Anti-migration Properties on Androgen-independent Human Prostate Cancer by Targeting Cell Cycle-Apoptosis and PI3K Signalling Pathways. Front Pharmacol 2021; 12:707335. [PMID: 34366863 PMCID: PMC8343533 DOI: 10.3389/fphar.2021.707335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/02/2021] [Indexed: 01/10/2023] Open
Abstract
Diarylpentanoids exhibit a high degree of anti-cancer activity and stability in vitro over curcumin in prostate cancer cells. Hence, this study aims to investigate the effects of a diarylpentanoid, 1,5-bis(4-hydroxy-3-methoxyphenyl)-1,4-pentadiene-3-one (MS13) on cytotoxicity, anti-proliferative, apoptosis-inducing, anti-migration properties, and the underlying molecular mechanisms on treated androgen-independent prostate cancer cells, DU 145 and PC-3. A cell viability assay has shown greater cytotoxicity effects of MS13-treated DU 145 cells (EC50 7.57 ± 0.2 µM) and PC-3 cells (EC50 7.80 ± 0.7 µM) compared to curcumin (EC50: DU 145; 34.25 ± 2.7 µM and PC-3; 27.77 ± 6.4 µM). In addition, MS13 exhibited significant anti-proliferative activity against AIPC cells compared to curcumin in a dose- and time-dependent manner. Morphological observation, increased caspase-3 activity, and reduced Bcl-2 protein levels in these cells indicated that MS13 induces apoptosis in a time- and dose-dependent. Moreover, MS13 effectively inhibited the migration of DU 145 and PC-3 cells. Our results suggest that cell cycle-apoptosis and PI3K pathways were the topmost significant pathways impacted by MS13 activity. Our findings suggest that MS13 may demonstrate the anti-cancer activity by modulating DEGs associated with the cell cycle-apoptosis and PI3K pathways, thus inhibiting cell proliferation and cell migration as well as inducing apoptosis in AIPC cells.
Collapse
Affiliation(s)
- Nurul Azwa Abd Wahab
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, Serdang, Malaysia.,Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
16
|
Fu X, Cui J, Meng X, Jiang P, Zheng Q, Zhao W, Chen X. Endoplasmic reticulum stress, cell death and tumor: Association between endoplasmic reticulum stress and the apoptosis pathway in tumors (Review). Oncol Rep 2021; 45:801-808. [PMID: 33469681 PMCID: PMC7859917 DOI: 10.3892/or.2021.7933] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
External and internal stimuli are often involved in the pathogenesis of tumors, and the deterioration of endoplasmic reticulum (ER) function within cells is also an important etiological factor of tumorigenesis resulting in the impairment of the endoplasmic reticulum, which is termed ER stress. The ER is an organelle that serves a crucial role in the process of protein synthesis and maturation, and also acts as a reservoir of calcium to maintain intracellular Ca2+ homeostasis. ER stress has been revealed to serve a critical role in tumorigenesis. In the present review, the association between ER stress‑related pathways and tumor cell apoptosis is examined. Primarily, the role of ER stress in tumor cell apoptosis is discussed, and it is stipulated that ER stress, induced by drugs both directly and indirectly, promotes tumor cell apoptosis.
Collapse
Affiliation(s)
- Xiaojing Fu
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Juanjuan Cui
- Qingdao Municipal Hospital, Qingdao (Group), Qingdao, Shandong 266071, P.R. China
| | - Xiangjun Meng
- Qingdao Mental Health Center, Qingdao, Shandong 266071, P.R. China
| | - Piyu Jiang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Qiuling Zheng
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Wenwen Zhao
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xuehong Chen
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
17
|
|
18
|
Li Z, He P, Luo G, Shi X, Yuan G, Zhang B, Seidl C, Gewies A, Wang Y, Zou Y, Long Y, Yue D, Zhang X. Increased Tumoral Microenvironmental pH Improves Cytotoxic Effect of Pharmacologic Ascorbic Acid in Castration-Resistant Prostate Cancer Cells. Front Pharmacol 2020; 11:570939. [PMID: 33071784 PMCID: PMC7538777 DOI: 10.3389/fphar.2020.570939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/31/2020] [Indexed: 01/23/2023] Open
Abstract
Background The anticancer potential of pharmacologic ascorbic acid (AA) has been detected in a number of cancer cells. However, in vivo study suggested a strongly reduced cytotoxic activity of AA. It was known that pH could be a critical influencing factor for multiple anticancer treatments. In this study, we explored the influence of pH on the cytotoxicity of ascorbic acid. We employed castration-resistant prostate cancer (CRPC) cell lines PC3 and DU145 to observe the therapeutic effect of AA on PCa cells that were cultured with different pH in vitro. We also analyzed the influence of pH and extracellular oxidation on cytotoxicity of AA in cancer cells using reactive oxygen species (ROS) assay, cellular uptake of AA, and NADPH assay. Male BALB/c nude mice bearing prostate carcinoma xenografts (PC3 or DU145) were used to assess treatment response to AA with or without bicarbonate in vivo. The cellular uptake of AA in PCa xenografts was detected using positron emission tomography (PET). Small animal PET/CT scans were performed on mice after the administration of 6-deoxy-6-[18F] fluoro-L-ascorbic acid (18F-DFA). Results Our in vitro studies demonstrate that acidic pH attenuates the cytotoxic activity of pharmacologic ascorbic acid by inhibiting AA uptake in PCa cells. Additionally, we found that the cancer cell-selective toxicity of AA depends on ROS. In vivo, combination of AA and bicarbonate could provide a significant better therapeutic outcome in comparison with controls or AA single treated mice. 18F-DFA PET imaging illustrated that the treatment with NaHCO3 could significantly increase the AA uptake in tumor. Conclusions The alkalinity of tumor microenvironment plays an important role in anticancer efficiency of AA in CRPC. 18F-DFA PET/CT imaging could predict the therapeutic response of PCa animal model through illustration of tumoral uptake of AA. 18F-DFA might be a potential PET tracer in clinical diagnosis and treatment for CRPC.
Collapse
Affiliation(s)
- Zhoulei Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Peng He
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ganhua Luo
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xinchong Shi
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Gang Yuan
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bing Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Christof Seidl
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Andreas Gewies
- Institute of Molecular Toxicology and Pharmacology, German Research Center for Environmental Health, Munich, Germany
| | - Yue Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuan Zou
- Sichuan Key Laboratory of Medical Imaging & Ultrasound Medical Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yali Long
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dianchao Yue
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiangsong Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
19
|
The curcumin analogue WZ35 affects glycolysis inhibition of gastric cancer cells through ROS-YAP-JNK pathway. Food Chem Toxicol 2020; 137:111131. [DOI: 10.1016/j.fct.2020.111131] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/11/2022]
|
20
|
Leong SW, Chia SL, Abas F, Yusoff K. Synthesis and in-vitro anti-cancer evaluations of multi-methoxylated asymmetrical diarylpentanoids as intrinsic apoptosis inducer against colorectal cancer. Bioorg Med Chem Lett 2020; 30:127065. [PMID: 32127259 DOI: 10.1016/j.bmcl.2020.127065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 01/13/2023]
Abstract
In the present study, a series of nine stable 3,4,5-methoxylphenyl-containing asymmetrical diarylpentanoids, derivatives of curcuminoids, have been synthesized, characterized and evaluated for their in-vitro anti-cancer potential against a panel of BRAF- and KRAS-mutated colorectal cancer cell lines including T84, LoVo and SW620, HT29, RKO and NCI-H508, respectively. Structure-activity relationship study on cytotoxicity of tested compounds suggested that the presence of meta-hydroxyl and adjacent dimethoxyl groups are crucial for enhanced cytotoxicity of diarylpentanoids. Among the evaluated analogs, 8 has been identified as the lead compound due to its highest chemotherapeutic index of 9.9 and nano molar scale cytotoxicity against SW620 and RKO. Colonies formation and cell cycle analyses on 8-treated RKO cells showed that 8 exhibits strong anti-proliferative activity by inducing G2/M-phase cell arrest. Subsequent flow cytometry based annexin-V and DCFHDA studies suggested that 8 could induce apoptosis through intracellular ROS-dependent pathway. Further Western blot studies confirmed that 8 has induced intrinsic apoptosis in RKO cells through the up-regulations of Bad and Bax pro-apoptotic proteins and down-regulations of Bcl-2 and Bcl-xL pro-survival proteins. In all, the present results suggest that 8 could be a potent lead which deserves further modification and investigation in the development of small molecule-based anti-colorectal cancer agents.
Collapse
Affiliation(s)
- Sze Wei Leong
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia.
| | - Suet Lin Chia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia; Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia.
| | - Faridah Abas
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia; Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor Darul Ehsan, Malaysia; Malaysia Genome Institute (MGI), National Institute of Biotechnology Malaysia (NIBM), Jalan Bangi, 43000 Kajang, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
21
|
Stępień K, Wojdyła D, Nowak K, Mołoń M. Impact of curcumin on replicative and chronological aging in the Saccharomyces cerevisiae yeast. Biogerontology 2020; 21:109-123. [PMID: 31659616 PMCID: PMC6942599 DOI: 10.1007/s10522-019-09846-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022]
Abstract
Curcumin is a biologically active compound of vegetable origin which has a hormetic effect. Pro-health and anti-aging properties of curcumin have been known for years. The main benefit of curcumin is thought to be its anti-oxidative action. Despite vast amount of data confirming age-delaying activity of curcumin in various groups of organisms, so far little has been discovered about curcumin's impact on cell aging in the experimental model of the Saccharomyces cerevisiae budding yeast. We have been able to demonstrate that curcumin significantly increases oxidative stress and accelerates replicative and chronological aging of yeast cells devoid of anti-oxidative protection (with SOD1 and SOD2 gene deletion) and deprived of DNA repair mechanisms (RAD52). Interestingly, curcumin delays aging, probably through hormesis, of the wild-type strain BY4741.
Collapse
Affiliation(s)
- Karolina Stępień
- Department of Biochemistry and Cell Biology, University of Rzeszow, Zelwerowicza 4, 35-601, Rzeszow, Poland
| | - Dominik Wojdyła
- Department of Biochemistry and Cell Biology, University of Rzeszow, Zelwerowicza 4, 35-601, Rzeszow, Poland
| | - Katarzyna Nowak
- Department of Biochemistry and Cell Biology, University of Rzeszow, Zelwerowicza 4, 35-601, Rzeszow, Poland
| | - Mateusz Mołoń
- Department of Biochemistry and Cell Biology, University of Rzeszow, Zelwerowicza 4, 35-601, Rzeszow, Poland.
| |
Collapse
|
22
|
Rajamanickam V, Yan T, Wu L, Zhao Y, Xu X, Zhu H, Chen X, Wang M, Liu Z, Liu Z, Liang G, Wang Y. Allylated Curcumin Analog CA6 Inhibits TrxR1 and Leads to ROS-Dependent Apoptotic Cell Death in Gastric Cancer Through Akt-FoxO3a. Cancer Manag Res 2020; 12:247-263. [PMID: 32021440 PMCID: PMC6968823 DOI: 10.2147/cmar.s227415] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022] Open
Abstract
Background Gastric cancer is one of the leading causes of cancer-related deaths. Allylated monocarbonyl analogs of curcumin (MACs) have been reported to selectively inhibit a broad range of human cancers including gastric cancer. However, the precise molecular mechanisms underlying the inhibitory activities of MACs are not fully known. Methods In this study, we examined the anti-tumor activities of an allylated MAC, CA6, on gastric cancer cells and gastric cancer xenograft mouse model. The potential molecular anti-tumor mechanisms of CA6 were also elucidated. Results Our data show that CA6 exhibited significant cytotoxicity in gastric cancer cells, which was seen as an induction of G2/M cell cycle arrest and apoptosis. These activities were mediated through an elaboration of ROS levels in gastric cancer cells and induction of endoplasmic reticulum stress. CA6 increased ROS levels through directly binding to and inhibiting thioredoxin reductase R1 (TrxR1). Also, CA6-generated ROS inhibited Akt and activated forkhead O3A (FoxO3a), causing cytotoxicity in gastric cancer cells. Finally, CA6 treatment dose-dependently reduced the growth of gastric cancer xenografts in tumor-bearing mice, which was associated with reduced TrxR1 activity and increased ROS in the tumor. Conclusion In summary, our studies demonstrate that CA6 inhibited gastric cancer growth by inhibiting TrxR1 and increasing ROS, which in turn activated FoxO3a through suppressing Akt. CA6 is a potential candidate for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Vinothkumar Rajamanickam
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Tao Yan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Liangrong Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yanni Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Xiaohong Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Heping Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Xi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Meihong Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Zhoudi Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Zhiguo Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| |
Collapse
|
23
|
Curcumin and related antioxidants: applications to tissue pathology. Pathology 2020. [DOI: 10.1016/b978-0-12-815972-9.00019-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
24
|
Xiang DB, Zhang KQ, Zeng YL, Yan QZ, Shi Z, Tuo QH, Lin LM, Xia BH, Wu P, Liao DF. Curcumin: From a controversial "panacea" to effective antineoplastic products. Medicine (Baltimore) 2020; 99:e18467. [PMID: 31914018 PMCID: PMC6959860 DOI: 10.1097/md.0000000000018467] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Curcumin, a controversial "panacea," has been broadly studied. Its bioactivities including antioxidant, anti-inflammatory, and especially antineoplastic activities have been documented. However, due to its extensive bioactivities, some scientists hold a skeptical point of view toward curcumin and described curcumin as a "deceiver" to chemists. The objective of this study was to explore curcumin's another possibility as a potential supplementary leading compound to cancer treatments. METHODS Literature searches were conducted using electronic databases. Search terms such as "curcumin," "curcumin analogues," and so on were used. The literatures were collected and summarized. In this article, reported targets of curcumin are reviewed. The limitations of a curcumin as a therapeutic anticancer product including low bioavailability and poor targeting are mentioned. Furthermore, modified curcumin analogues and antitumor mechanisms are listed and discussed in the aspects of cell death and tumor microenvironment including angiogenesis, tissue hypoxia status, and energy metabolism. RESULTS Several possible modification strategies were presented by analyzing the relationships between the antitumor activity of curcumin analogues and their structural characteristics, including the introduction of hydrophilic group, shortening of redundant hydrocarbon chain, the introduction of extra chemical group, and so on. CONCLUSIONS From our perspective, after structural modification curcumin could be more effective complementary product for cancer therapies by the enhancement of targeting abilities and the improvement of bioavailability.
Collapse
Affiliation(s)
- De-Biao Xiang
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Kai-Qiang Zhang
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Ya-Ling Zeng
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qing-Zi Yan
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Zhe Shi
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Qin-Hui Tuo
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Li-Mei Lin
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Bo-Hou Xia
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Ping Wu
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| |
Collapse
|
25
|
Subramaniam S, Selvaduray KR, Radhakrishnan AK. Bioactive Compounds: Natural Defense Against Cancer? Biomolecules 2019; 9:biom9120758. [PMID: 31766399 PMCID: PMC6995630 DOI: 10.3390/biom9120758] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer is a devastating disease that has claimed many lives. Natural bioactive agents from plants are gaining wide attention for their anticancer activities. Several studies have found that natural plant-based bioactive compounds can enhance the efficacy of chemotherapy, and in some cases ameliorate some of the side-effects of drugs used as chemotherapeutic agents. In this paper, we have reviewed the literature on the anticancer effects of four plant-based bioactive compounds namely, curcumin, myricetin, geraniin and tocotrienols (T3) to provide an overview on some of the key findings that are related to this effect. The molecular mechanisms through which the active compounds may exert their anticancer properties in cell and animal-based studies also discussed.
Collapse
Affiliation(s)
- Shonia Subramaniam
- Pathology Division, School of Medicine, International Medical University, Bukit Jalil, Kuala Lumpur 50050, Malaysia;
- Product Development and Advisory Services, Malaysian Palm Oil Board, Kajang, Selangor 43000, Malaysia;
| | - Kanga Rani Selvaduray
- Product Development and Advisory Services, Malaysian Palm Oil Board, Kajang, Selangor 43000, Malaysia;
| | - Ammu Kutty Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
- Correspondence: ; Tel.: +60-355-144-902
| |
Collapse
|
26
|
Insights on the synthesis of asymmetric curcumin derivatives and their biological activities. Eur J Med Chem 2019; 183:111704. [PMID: 31557608 DOI: 10.1016/j.ejmech.2019.111704] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/02/2019] [Accepted: 09/14/2019] [Indexed: 02/08/2023]
Abstract
Curcumin is a small organic molecule with pleiotropic biological activities. However, its multiple structural-pharmacokinetic challenges prevent its development into a clinical drug. Various structural modifications have been made to improve its drug profile. In this review, we focus on the methods adopted in the synthesis of asymmetric curcumin derivatives and their biological activities and forecast the future of this exciting class of compounds in the field of medicine.
Collapse
|
27
|
Dual role of Endoplasmic Reticulum Stress-Mediated Unfolded Protein Response Signaling Pathway in Carcinogenesis. Int J Mol Sci 2019; 20:ijms20184354. [PMID: 31491919 PMCID: PMC6770252 DOI: 10.3390/ijms20184354] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer constitutes a grave problem nowadays in view of the fact that it has become one of the main causes of death worldwide. Poor clinical prognosis is presumably due to cancer cells metabolism as tumor microenvironment is affected by oxidative stress. This event triggers adequate cellular response and thereby creates appropriate conditions for further cancer progression. Endoplasmic reticulum (ER) stress occurs when the balance between an ability of the ER to fold and transfer proteins and the degradation of the misfolded ones become distorted. Since ER is an organelle relatively sensitive to oxidative damage, aforementioned conditions swiftly cause the activation of the unfolded protein response (UPR) signaling pathway. The output of the UPR, depending on numerous factors, may vary and switch between the pro-survival and the pro-apoptotic branch, and hence it displays opposing effects in deciding the fate of the cancer cell. The role of UPR-related proteins in tumorigenesis, such as binding the immunoglobulin protein (BiP) and inositol-requiring enzyme-1α (IRE1α), activating transcription factor 6 (ATF6) or the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), has already been specifically described so far. Nevertheless, due to the paradoxical outcomes of the UPR activation as well as gaps in current knowledge, it still needs to be further investigated. Herein we would like to elicit the actual link between neoplastic diseases and the UPR signaling pathway, considering its major branches and discussing its potential use in the development of a novel, anti-cancer, targeted therapy.
Collapse
|
28
|
Shakeri A, Zirak MR, Wallace Hayes A, Reiter R, Karimi G. Curcumin and its analogues protect from endoplasmic reticulum stress: Mechanisms and pathways. Pharmacol Res 2019; 146:104335. [DOI: 10.1016/j.phrs.2019.104335] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023]
|
29
|
Hu X, Zheng W, Luo Y, Ou X, Song L, Zhang S, He T, Guo Z, Zhu J, Shi H, Huang W, Yu R. Arca subcrenata Polypeptides Inhibit Human Colorectal Cancer HT-29 Cells Growth via Suppression of IGF-1R/Akt/mTOR Signaling and ATP Production. Nutr Cancer 2019; 72:260-272. [PMID: 31295033 DOI: 10.1080/01635581.2019.1625935] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Arca subcrenata Lischke, widely scattering offshore at neritic regions, is very popular on dining table due to its edible and medical functional meatball. This study aims to investigate the suppression of a polypeptide fraction from A. subcrenata (PAS) on human colorectal cancer HT-29 cells, and its underlying mechanism. The results showed that PAS inhibited the growth of HT-29 cells with an IC50 value of 117 μg/ml after 48 h treatment, and significantly suppressed the tumor growth in nude mice bearing-xenografted HT-29 cells at the dosage of 63 mg/kg, with little influence on normal colon cells and normal colonic mucosa. PAS was then inspiringly found to induce apoptosis and G2/M phase arrest in HT-29 cells. The effect mechanism was involved in the inhibition of IGF-1/IGF-1R signaling activation, which was responsible for inactivating downstream Akt/mTOR pathway. Immunofluorescence assay also showed that PAS could reduce phosphorylation of IGF-1R (Tyr1165/1166). IGF-1, an IGF-1R activator, could reverse the suppression of PAS on IGF-1R phosphorylation. Furthermore, PAS significantly inhibited ATP production of HT-29 cells both in vitro and in vivo. Our results provide positive evidence that A. subcrenata has the potential to be a candidate for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Xianjing Hu
- Biotechnological Institute of Chinese Materia Medical, Jinan University, Guangzhou, China.,Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Weiming Zheng
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yuanyuan Luo
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaozheng Ou
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Liyan Song
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Sirui Zhang
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Tingsha He
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhongyi Guo
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jianhua Zhu
- Biotechnological Institute of Chinese Materia Medical, Jinan University, Guangzhou, China
| | - Hui Shi
- Biotechnological Institute of Chinese Materia Medical, Jinan University, Guangzhou, China
| | - Weijuan Huang
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| | - Rongmin Yu
- Biotechnological Institute of Chinese Materia Medical, Jinan University, Guangzhou, China.,Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, China
| |
Collapse
|
30
|
Araveti PB, Srivastava A. Curcumin induced oxidative stress causes autophagy and apoptosis in bovine leucocytes transformed by Theileria annulata. Cell Death Discov 2019; 5:100. [PMID: 31231548 PMCID: PMC6547749 DOI: 10.1038/s41420-019-0180-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/17/2019] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Bovine tropical theileriosis is a tick-borne disease, caused by Theileria annulata which is a protozoan parasite that resides within the B-cells and macrophages. T. annulata is a unique parasite that can transform bovine leucocytes which leads to the cancer hallmarks in the infected cells. Previously, curcumin has been shown to possess multiple pharmacological activities such as anti-inflammatory and anti-cancer activities. In this study, we demonstrated that curcumin inhibits the proliferation of Theileria-transformed bovine leucocytes by promoting apoptosis and autophagy. The transcriptome analysis of curcumin treated cells showed that the genes involved in cell death and autophagy are also differentially regulated. We further elucidated the mechanism of action of curcumin on Theileria infected bovine cells. We found that curcumin induced the generation of reactive oxygen species (ROS) which activated caspase 8 and destabilized the mitochondrial membrane potential leading to the release of cytochrome c from mitochondria. This subsequently led to the activation of caspase 3 and PARP cleavage, finally leading to apoptosis in the infected cells. Furthermore, curcumin induced the process of autophagy which was characterized by the formation of acidic vesicular organelles, LC3B accumulation with lysosome inhibitor, E64d, and the presence of autophagosomes as visualized by transmission electron microscopy (TEM). Curcumin treatment suppressed the mTOR and increased the expression of autophagy-related proteins. We also found that N- acetylcysteine, an inhibitor of ROS, could rescue the infected cells from curcumin induced apoptosis and autophagy mediated cell death. Intriguingly, curcumin had no effect on uninfected bovine PBMCs. Altogether, these data suggest the therapeutic potential of curcumin against bovine tropical theileriosis.
Collapse
Affiliation(s)
| | - Anand Srivastava
- National Institute of Animal Biotechnology (NIAB), Hyderabad, India
| |
Collapse
|
31
|
Rodrigues FC, Anil Kumar NV, Thakur G. Developments in the anticancer activity of structurally modified curcumin: An up-to-date review. Eur J Med Chem 2019; 177:76-104. [PMID: 31129455 DOI: 10.1016/j.ejmech.2019.04.058] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/14/2019] [Accepted: 04/20/2019] [Indexed: 01/10/2023]
Abstract
Curcumin is a pharmacologically active polyphenol derived from the popular spice element-Turmeric. The therapeutic activity of curcumin has been extensively investigated over the last few decades and reports suggest the role of curcumin in a large number of biological activities, particularly its prominent anticancer activity. Curcumin, being a pleiotropic molecule, is a regulator of multiple molecular targets which play crucial roles in various cell signaling pathways. It is known to suppress transformation, inhibit proliferation as well as induce apoptosis. However, despite all these benefits, the efficacy of curcumin remains limited due to its poor bioavailability, poor absorption within the systemic circulation and rapid elimination from the body. To overcome these limiting factors, researchers all around the world are working towards designing a synthetic and superior curcuminoid by making suitable structural modifications to the parent skeleton. These curcuminoids, mainly analogues and derivatives, will not only improve the physicochemical properties but also enhance the efficacy simultaneously. The present review will provide a comprehensive account of the analogues and derivatives of curcumin that have been reported since 2014 which have indicated a better anticancer activity than curcumin.
Collapse
Affiliation(s)
- Fiona C Rodrigues
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, India
| | - N V Anil Kumar
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, India
| | - Goutam Thakur
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, India.
| |
Collapse
|
32
|
Sea Cucumber ( Stichopus japonicas) F2 Enhanced TRAIL-Induced Apoptosis via XIAP Ubiquitination and ER Stress in Colorectal Cancer Cells. Nutrients 2019; 11:nu11051061. [PMID: 31083595 PMCID: PMC6567290 DOI: 10.3390/nu11051061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 05/08/2019] [Indexed: 01/16/2023] Open
Abstract
Natural products have shown great promise in sensitizing cells to TNF-related apoptosis-inducing ligand (TRAIL) therapy. Sea cucumber (SC) extracts possess antitumor activity, and hence their potential to sensitize colorectal cancer (CRC) cells to TRAIL therapy was evaluated. This study used Western blotting to evaluate the combination effects of SC and TRAIL in CRC, and determined the molecular mechanism underlying these effects. SC fractions and TRAIL alone did not affect apoptosis; however, combined treatment dramatically induced the apoptosis of CRC cells, but not of normal colon cells. Combined treatment induced the expression of apoptotic proteins (poly (ADP-ribose) polymerase (PARP), caspase 3, and 8), and this effect was markedly inhibited by the ubiquitination of X-linked inhibitor of apoptosis protein (XIAP). SC did not affect the mRNA levels, but it increased proteasomal degradation and ubiquitination of the XIAP protein. Furthermore, SC induced reactive oxygen species (ROS) production, thereby activating c-Jun N-terminal kinase (JNK) and endoplasmic reticulum (ER) stress-related apoptotic pathways in CRC. Altogether, our results demonstrate that the SC F2 fraction may sensitize CRC cells to TRAIL-induced apoptosis through XIAP ubiquitination and ER stress.
Collapse
|
33
|
Bloom Syndrome Protein Activates AKT and PRAS40 in Prostate Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3685817. [PMID: 31210839 PMCID: PMC6532288 DOI: 10.1155/2019/3685817] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/07/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
Abstract
Purpose Prostate cancer (PC) is a common malignant tumor and a leading cause of cancer-related death in men worldwide. In order to design new therapeutic interventions for PC, an understanding of the molecular events underlying PC tumorigenesis is required. Bloom syndrome protein (BLM) is a RecQ-like helicase, which helps maintain genetic stability. BLM dysfunction has been implicated in tumor development, most recently during PC tumorigenesis. However, the molecular basis for BLM-induced PC progression remains poorly characterized. In this study, we investigated whether BLM modulates the phosphorylation of an array of prooncogenic signaling pathways to promote PC progression. Methods We analyzed differentially expressed proteins (DEPs) using iTRAQ technology. Site-directed knockout of BLM in PC-3 prostate cancer cells was performed using CRISPR/Cas9-mediated homologous recombination gene editing to confirm the effects of BLM on DEPs. PathScan® Antibody Array Kits were used to analyze the phosphorylation of nodal proteins in PC tissue. Immunohistochemistry and automated western blot (WES) analyses were used to validate these findings. Results We found that silencing BLM in PC-3 cells significantly reduced their proliferative capacity. In addition, BLM downregulation significantly reduced levels of phosphorylated protein kinase B (AKT (Ser473)) and proline-rich AKT substrate of 40 kDa (PRAS40 (Thr246)), and this was accompanied by enhanced ROS (reactive oxygen species) levels. In addition, we found that AKT and PRAS40 inhibition reduced BLM, increased ROS levels, and induced PC cell apoptosis. Conclusions We demonstrated that BLM activates AKT and PRAS40 to promote PC cell proliferation and survival. We further propose that ROS act in concert with BLM to facilitate PC oncogenesis, potentially via further enhancing AKT signaling and downregulating PTEN expression. Importantly, inhibiting the BLM-AKT-PRAS40 axis induced PC cell apoptosis. Thus, we highlight new avenues for novel anti-PC treatments.
Collapse
|
34
|
Sun Y, Liu L, Wang Y, He A, Hu H, Zhang J, Han M, Huang Y. Curcumin inhibits the proliferation and invasion of MG-63 cells through inactivation of the p-JAK2/p-STAT3 pathway. Onco Targets Ther 2019; 12:2011-2021. [PMID: 30936718 PMCID: PMC6421868 DOI: 10.2147/ott.s172909] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose The aims of this study were to determine the effect of curcumin on osteosarcoma (OS) cells due to inactivation of the p-JAK2/p-STAT3 pathway and evaluate the prognostic value of this pathway in OS. Materials and methods We exposed a human OS cell line (MG-63) to different concentrations of curcumin. Then, we characterized the effects on MG-63 cells using assays (cell viability, colony formation, cell cycle, wound healing, invasion), flow cytometry, Western blot, immunohistochemical analyses, and tumor xenograft. Results The half-maximal inhibitory of curcumin for MG-63 cells at 24 hours was 27.6 µM. The number of colonies of MG-63 cells was decreased obviously upon curcumin (10 and 20 µM) treatment. We also found increased accumulation of MG-63 cells in the G2/M phase upon curcumin (10 and 20 µM) treatment. Apoptosis was increased in 10 and 20 µM curcumin-treated MG-63 cells. After incubation of physically wounded cells for 24 hours, the percentage wound width increased upon curcumin exposure. Curcumin obviously decreased the expression of pJAK-2 and pSTAT-3 in MG-63 cells in a dose-dependent manner. Curcumin dose-dependently inhibited the proliferation, migration, and invasion of MG-63 cells and induced arrest of the G0/G1 phase and apoptosis by inhibiting the p-JAK2/p-STAT3 pathway. The linear correlativity between expression of p-JAK2 and STAT3 was very prominent, and both were closely associated with lung metastasis. In vivo study suggested that curcumin suppressed tumor growth through JAK2/STAT3 signaling. Conclusion Curcumin-mediated inhibition of the proliferation and migration of MG-63 cells was associated with inactivation of JAK/STAT signaling.
Collapse
Affiliation(s)
- Yuanjue Sun
- Cancer Therapy and Research Center, Shandong Provincial Hospital, Shandong University, 250021, Shandong, People's Republic of China,
| | - Liguo Liu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, People's Republic of China
| | - Yaling Wang
- Department of Medical Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, People's Republic of China,
| | - Aina He
- Department of Medical Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, People's Republic of China,
| | - Haiyan Hu
- Department of Medical Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, People's Republic of China,
| | - Jianjun Zhang
- Department of Medical Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, People's Republic of China,
| | - Mingyong Han
- Cancer Therapy and Research Center, Shandong Provincial Hospital, Shandong University, 250021, Shandong, People's Republic of China,
| | - Yujing Huang
- Department of Medical Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, People's Republic of China,
| |
Collapse
|
35
|
Wang C, Song X, Shang M, Zou W, Zhang M, Wei H, Shao H. Curcumin exerts cytotoxicity dependent on reactive oxygen species accumulation in non-small-cell lung cancer cells. Future Oncol 2019; 15:1243-1253. [PMID: 30843426 DOI: 10.2217/fon-2018-0708] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM Curcumin induces cytotoxic cell death in several human cancer cells. Here, we have investigated the effects of curcumin on non-small-cell lung cancer (NSCLC) with an aim to identify underlying mechanisms of its cytotoxic effect. MATERIALS & METHODS The effects of various concentrations of curcumin on the NSCLC cell lines A549 and SPC-A1 were evaluated by MTT assay, colony-forming assay and flow cytometry. Additionally, protein expression associated with different signaling pathways was assessed using western blotting. RESULTS Curcumin exhibited cytotoxicity against NSCLC, evident from the inhibition of cell proliferation, G2/M arrest, DNA damage, endoplasmic reticulum stress and mitochondrial apoptosis. The anticancer effect was related to reactive oxygen species (ROS) accumulation and could be reversed by ROS scavengers, catalase and N-acetyl-l-cysteine. Curcumin decreased mitochondrial transmembrane potential and induced ROS production, thereby activating the DNA damage/repair pathway and mitochondrial apoptosis. CONCLUSION These results indicate that curcumin could be an effective therapeutic candidate for NSCLC.
Collapse
Affiliation(s)
- Cuijuan Wang
- Key Laboratory of Public Health, Shandong Academy of Occupational Health & Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Xingguo Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Ming Shang
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Wei Zou
- Key Laboratory of Public Health, Shandong Academy of Occupational Health & Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Mengping Zhang
- Key Laboratory of Public Health, Shandong Academy of Occupational Health & Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Haiyan Wei
- Key Laboratory of Public Health, Shandong Academy of Occupational Health & Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| | - Hua Shao
- Key Laboratory of Public Health, Shandong Academy of Occupational Health & Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, PR China
| |
Collapse
|
36
|
Lee ES, Kwon MH, Kim HM, Kim N, Kim YM, Kim HS, Lee EY, Chung CH. Dibenzoylmethane ameliorates lipid-induced inflammation and oxidative injury in diabetic nephropathy. J Endocrinol 2019; 240:169-179. [PMID: 30475214 DOI: 10.1530/joe-18-0206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/31/2018] [Indexed: 11/08/2022]
Abstract
Dibenzoylmethane (DBM) is a beta-diketone analog of curcumin. Numerous studies have shown the beneficial effects of curcumin on diabetes, obesity and diabetic complications including diabetic nephropathy. Recently, we investigated the beneficial metabolic effects of DBM on high-fat diet-induced obesity. However, the effects and mechanisms of action of DBM in the kidney are currently unknown. To investigate the renoprotective effects of DBM in type 2 diabetes, we administered DBM (100 mg/kg) orally for 12 weeks to high-fat diet-induced diabetic model mice. We used mouse renal mesangial (MES13) and macrophage (RAW 264.7) cells to examine the mechanism of action of DBM (20 μM). After DBM treatment, the albumin-to-creatinine ratio was significantly decreased compared to that of the high-fat-diet group. Moreover, damaged renal ultra-structures and functions including increased glomerular volume, glomerular basement membrane thickness and inflammatory signals were ameliorated after DBM treatment. Stimulation of MES13 and RAW264.7 cells by palmitate or high-dose glucose with lipopolysaccharides increased inflammatory signals and macrophage migration. However, these changes were reversed by DBM treatment. In addition, DBM inhibited NADPH oxidase 2 and 4 expression and oxidative DNA damage. Collectively, these data suggested that DBM prevented diabetes-induced renal injury through its anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Eun Soo Lee
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Mi-Hye Kwon
- The East Coast Research Institute of Life Science, Gangneung-Wonju National University, Gangneung, Korea
| | - Hong Min Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Nami Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - You Mi Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hyeon Soo Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | - Eun Young Lee
- Department of Internal Medicine and Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Korea
| | - Choon Hee Chung
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
37
|
Wang L, Zhu Z, Han L, Zhao L, Weng J, Yang H, Wu S, Chen K, Wu L, Chen T. A curcumin derivative, WZ35, suppresses hepatocellular cancer cell growthviadownregulating YAP-mediated autophagy. Food Funct 2019; 10:3748-3757. [PMID: 31172987 DOI: 10.1039/c8fo02448k] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
HCC is a common cancer type in the world. Here, we found WZ35, a novel derivative of curcumin, could notably suppress HCC cell growthviainhibiting YAP controlled autophagy, highlighting the potent anti-tumor activity of WZ35 in liver cancer therapy.
Collapse
Affiliation(s)
- Lihua Wang
- School of Ophthalmology and Optometry
- Eye Hospital
- Wenzhou Medical University
- Wenzhou 325000
- China
| | - Zheng Zhu
- Laboratory Animal Centre
- Wenzhou Medical University
- Wenzhou
- China
| | - Lei Han
- Laboratory Animal Centre
- Wenzhou Medical University
- Wenzhou
- China
| | - Liqian Zhao
- Laboratory Animal Centre
- Wenzhou Medical University
- Wenzhou
- China
| | - Jialei Weng
- Laboratory Animal Centre
- Wenzhou Medical University
- Wenzhou
- China
| | - Hongbao Yang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Shijia Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Kaiyuan Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Liang Wu
- Department of Pathology
- First Affiliated Hospital of WenZhou Medical University
- Wenzhou 325000
- China
| | - Tongke Chen
- Laboratory Animal Centre
- Wenzhou Medical University
- Wenzhou
- China
| |
Collapse
|
38
|
Li X, Wu J, Zhang X, Chen W. Glutathione reductase-mediated thiol oxidative stress suppresses metastasis of murine melanoma cells. Free Radic Biol Med 2018; 129:256-267. [PMID: 30086340 DOI: 10.1016/j.freeradbiomed.2018.07.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023]
Abstract
Malignant melanoma is a highly metastatic and life-threatening cancer. Reactive oxygen species (ROS) play important roles in cancer initiation and progression including metastasis. It has been reported that the oxidative stress spontaneously generated in circulating melanoma cells was able to suppress distant metastasis in vivo. However, little is known regarding the effects and mechanism of glutathione reductase (GR) inhibition-induced oxidative stress in regulation of melanoma metastasis. Here, we demonstrate that GR inhibition generates oxidative stress and suppresses lung metastasis and subcutaneous growth of melanoma in vivo. In addition, inhibitory effects by GR activity reduction were observed on cell proliferation, colony formation, cell adhesion, migration and invasion in melanoma cells in vitro. GR inhibition-induced oxidative stress was also found to block epithelial-to-mesenchymal transition (EMT) by decreasing the expression of Vimentin, ERK1/2, transcription factor Snail and increasing the expression of E-cadherin. In addition, actin rearrangement, a key element involved in cell motility, was also affected by GR-mediated oxidative stress possibly through protein S-glutathionylation on actin. In conclusion, this study identifies GR as an effective regulator of oxidative stress that affects the multistep processes of metastasis in melanoma cells, and it becomes a potential target for melanoma therapy.
Collapse
Affiliation(s)
- Xia Li
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province 310022, China; Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province 310022, China
| | - Junzhou Wu
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province 310022, China
| | - Xiaoying Zhang
- ACEA Bio CO., Ltd., Hangzhou, Zhejiang Province 310030, China
| | - Wei Chen
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province 310022, China; Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province 310022, China.
| |
Collapse
|
39
|
Isobavachalcone Induces ROS-Mediated Apoptosis via Targeting Thioredoxin Reductase 1 in Human Prostate Cancer PC-3 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1915828. [PMID: 30410640 PMCID: PMC6206523 DOI: 10.1155/2018/1915828] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/02/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
Prostate carcinoma causes a great number of deaths every year; therefore, there is an urgent need to find new drug candidates to treat advanced prostate cancer. Isobavachalcone (IBC) is the chalcone composition of Psoralea corylifolia Linn used in traditional Chinese medicine. Although IBC demonstrates potent anticancer efficacy in numerous types of human cancer cells, the cellular targets of IBC have not been fully defined. In our study, we found that IBC may induce reactive oxygen species- (ROS-) mediated apoptosis via interaction with a selenocysteine (Sec) containing the antioxidant enzyme thioredoxin reductase 1 (TrxR1), and induce lethal endoplasmic reticulum (ER) stress by inhibiting TrxR1 activity and increasing ROS levels in human prostate cancer PC-3 cells. Furthermore, we also observed that knocking down TrxR1 would sensitized cancer cells to IBC treatment. Our study provides evidence for the anticancer mechanism of IBC with TrxR1 as a potential target.
Collapse
|
40
|
Wang L, Han L, Tao Z, Zhu Z, Han L, Yang Z, Wang H, Dai D, Wu L, Yuan Z, Chen T. The curcumin derivative WZ35 activates ROS-dependent JNK to suppress hepatocellular carcinoma metastasis. Food Funct 2018; 9:2970-2978. [PMID: 29766185 DOI: 10.1039/c8fo00314a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumor metastasis is the leading cause of cancer death; due to the progress made in the elucidation of the mechanism of cancer cell metastasis, there is hope for patients with severe stages of cancer. Curcumin, as a novel anti-cancer drug, has been applied in cancer therapy; however, the toxicity of curcumin hinders its application. Herein, we constructed a novel derivative, WZ35, and evaluated its metastatic inhibition properties in vitro and in vivo. CCK-8 assay was performed to evaluate the tumor suppressive activity of WZ35. Cell apoptosis was detected by flow cytometry analysis. Transwell cell migration assay and RTCA were used to detect cell migration in mock and WZ35-treated cells. Western blotting was performed to analyze molecular alteration with different treatments. In this study, we found that curcumin and its derivative WZ35 could dramatically suppress proliferation, invasion, and migration of the hepatocellular HCCLM3, HepG2, and Huh7 cancer cells. Moreover, the cancer cell metastatic markers MMP-2, MMP-9, and N-cadherin were decreased, and E-cadherin was up-regulated. In addition, our data show that WZ35 promotes ROS-dependent JNK activation that is essential for WZ35-caused cell metastasis suppression. Moreover, the NAC and JNK inhibitor SP600125 could dramatically reverse WZ35-caused MMP-2, MMP-9, and N-cadherin reduction and E-cadherin up-regulation. We have also found that WZ35 exhibits powerful anti-metastasis activity of HCCLM3 in vivo. In conclusion, our data indicated that WZ35 could be a candidate for the treatment of metastatic liver cancer patients.
Collapse
Affiliation(s)
- Lihua Wang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Dai X, Zhang J, Guo G, Cai Y, Cui R, Yin C, Liu W, Vinothkumar R, Zhang T, Liang G, Zhang X. A mono-carbonyl analog of curcumin induces apoptosis in drug-resistant EGFR-mutant lung cancer through the generation of oxidative stress and mitochondrial dysfunction. Cancer Manag Res 2018; 10:3069-3082. [PMID: 30214301 PMCID: PMC6124478 DOI: 10.2147/cmar.s159660] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Introduction Targeted therapies using epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) have revolutionized the treatment of non-small cell lung cancer (NSCLC) patients harboring EGFR mutations, leading to the approval of gefitinib and erlotinib as standard first-line clinical treatment. Inevitably, a considerable proportion of patients develop resistance to EGFR-TKIs due to the acquisition of secondary mutations within EGFR. Therefore, alternative strategies to target NSCLC are desperately needed. Materials and methods In this study, we have evaluated the effect of a reactive oxygen species (ROS)-inducing agent WZ35, a mono-carbonyl analog of curcumin, to target an inherent biological property of cancer cells, increased oxidative stress. To study whether WZ35 can inhibit NSCLC tumorigenesis, we used gefitinib- and erlotinib-resistant cell line H1975. Results In this study, we show that WZ35 treatment dramatically decreases cell viability and induces apoptosis in H1975 cells through the generation of ROS. We also found that the antitumor activity of WZ35 involved ROS-mediated activation of the endoplasmic reticulum stress pathway and mitochondrial dysfunction. Furthermore, WZ35 significantly inhibited H1975 xenograft tumor growth through the inhibition of cell proliferation and induction of apoptosis. Discussion These findings show that WZ35 may be a promising candidate for the treatment of EGFR-TKI-resistant NSCLC.
Collapse
Affiliation(s)
- Xuanxuan Dai
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Junru Zhang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Guilong Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Yuepiao Cai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Ri Cui
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Changtian Yin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Weidong Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Rajamanickam Vinothkumar
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Tingting Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Xiaohua Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| |
Collapse
|
42
|
Asymmetrical meta-methoxylated diarylpentanoids: Rational design, synthesis and anti-cancer evaluation in-vitro. Eur J Med Chem 2018; 157:716-728. [PMID: 30138803 DOI: 10.1016/j.ejmech.2018.08.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 08/11/2018] [Accepted: 08/14/2018] [Indexed: 12/18/2022]
Abstract
In the present study, a series of forty-five asymmetrical meta-methoxylated diarylpentanoids have been synthesized, characterized and evaluated for their in-vitro anti-cancer potential. Among the forty-five analogs, three compounds (20, 33 and 42) have been identified as lead compounds due to their excellent inhibition against five human cancer cell lines including SW620, A549, EJ28, HT1080 and MCF-7. Structure-activity relationship study on cytotoxicity of tested compounds suggested that the presence of meta-oxygenated phenyl ring played a critical role in enhancing their cytotoxic effects. Compounds 33 and 42 in particular, exhibited strongest cytotoxicity against tested cell lines with the IC50 values ranging from 1.1 to 4.3 μM. Subsequent colony formation assay on SW620 cell line showed that both compounds 33 and 42 possessed strong anti-proliferative activity. In addition, flow cytometry based experiments revealed that these compounds could trigger intracellular ROS production thus inducing G2/M-phase cell arrest and apoptosis. All these results suggested that poly meta-oxygenated diarylpentnoid is a promising scaffold which deserved further modification and investigation in the development of natural product-based anti-cancer drug.
Collapse
|
43
|
Liu H, Zhang X, Zhang S, Huang H, Wu J, Wang Y, Yuan L, Liu C, Zeng X, Cheng X, Zhuang D, Zhang H. Oxidative Stress Mediates Microcystin-LR-Induced Endoplasmic Reticulum Stress and Autophagy in KK-1 Cells and C57BL/6 Mice Ovaries. Front Physiol 2018; 9:1058. [PMID: 30131715 PMCID: PMC6090159 DOI: 10.3389/fphys.2018.01058] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/16/2018] [Indexed: 01/28/2023] Open
Abstract
Microcystin-leucine arginine (MC-LR) is a cyclic heptapeptide intracellular toxin released by cyanobacteria that exhibits strong reproductive toxicity. However, little is known about its biotoxicity to the female reproductive system. The present study investigates unexplored molecular pathways by which oxidative stress acts on MC-LR-induced endoplasmic reticulum stress (ERs) and autophagy. In the present study, immortalized murine ovarian granular cells (KK-1 cells) were exposed to 8.5, 17, and 34 μg/mL (IC50) of MC-LR with or without N-acetyl-l-cysteine (NAC, 10 mM) for 24 h, and C57BL/6 mice were treated with 12.5, 25.0, and 40.0 μg/kg⋅bw of MC-LR with or without NAC (200 mg/kg⋅bw) for 14 days. The results revealed that MC-LR could induce cells apoptosis and morphologic changes in ovarian tissues, induce oxidative stress by stimulating the generation of reactive oxygen species (ROS), destroying antioxidant capacity, and subsequently trigger ERs and autophagy by inducing the hyper-expression of ATG12, ATG5, ATG16, EIF2α (phosphorylated at S51), CHOP, XBP1, GRP78, Beclin1, and PERK (Thr980). Furthermore, NAC pretreatment partly inhibited MC-LR-induced ERs and autophagy via the PERK/ATG12 and XBP1/Beclin1 pathways. These results suggest that oxidative stress mediated MC-LR-induced ERs and autophagy in KK-1 cells and C57BL/6 mice ovaries. Therefore, oxidative stress plays an important role in female toxicity induced by MC-LR.
Collapse
Affiliation(s)
- Haohao Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Xiaofeng Zhang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Shenshen Zhang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Hui Huang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Jinxia Wu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yueqin Wang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Le Yuan
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Chuanrui Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Xin Zeng
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Xuemin Cheng
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Donggang Zhuang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Huizhen Zhang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
44
|
Theppawong A, Van de Walle T, Grootaert C, Bultinck M, Desmet T, Van Camp J, D'hooghe M. Synthesis of Novel Aza-aromatic Curcuminoids with Improved Biological Activities towards Various Cancer Cell Lines. ChemistryOpen 2018; 7:381-392. [PMID: 29872613 PMCID: PMC5974556 DOI: 10.1002/open.201800029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Indexed: 01/01/2023] Open
Abstract
Curcumin, a natural compound extracted from the rhizomes of Curcuma longa, displays pronounced anticancer properties but lacks good bioavailability and stability. In a previous study, we initiated structure modification of the curcumin scaffold by imination of the labile β-diketone moiety to produce novel β-enaminone derivatives. These compounds showed promising properties for elaborate follow-up studies. In this work, we focused on another class of nitrogen-containing curcuminoids with a similar objective: to address the bioavailability and stability issues and to improve the biological activity of curcumin. This paper thus reports on the synthesis of new pyridine-, indole-, and pyrrole-based curcumin analogues (aza-aromatic curcuminoids) and discusses their water solubility, antioxidant activity, and antiproliferative properties. In addition, multivariate statistics, including hierarchical clustering analysis and principal component analysis, were performed on a broad set of nitrogen-containing curcuminoids. Compared to their respective mother structures, that is, curcumin and bisdemethoxycurcumin, all compounds, and especially the pyridin-3-yl β-enaminone analogues, showed better water solubility profiles. Interestingly, the pyridine-, indole-, and pyrrole-based curcumin derivatives demonstrated improved biological effects in terms of mitochondrial activity impairment and protein content, in addition to comparable or decreased antioxidant properties. Overall, the biologically active N-alkyl β-enaminone aza-aromatic curcuminoids were shown to offer a desirable balance between good solubility and significant bioactivity.
Collapse
Affiliation(s)
- Atiruj Theppawong
- SynBioC Research GroupDepartment of Green Chemistry and TechnologyFaculty of Bioscience EngineeringGhent UniversityCoupure Links 6539000GhentBelgium
| | - Tim Van de Walle
- SynBioC Research GroupDepartment of Green Chemistry and TechnologyFaculty of Bioscience EngineeringGhent UniversityCoupure Links 6539000GhentBelgium
| | - Charlotte Grootaert
- Department of Food Technology, Safety and HealthFaculty of Bioscience EngineeringGhent UniversityCoupure Links 6539000GhentBelgium
| | - Margot Bultinck
- SynBioC Research GroupDepartment of Green Chemistry and TechnologyFaculty of Bioscience EngineeringGhent UniversityCoupure Links 6539000GhentBelgium
| | - Tom Desmet
- Department of BiotechnologyFaculty of Bioscience EngineeringGhent UniversityCoupure Links 6539000GhentBelgium
| | - John Van Camp
- Department of Food Technology, Safety and HealthFaculty of Bioscience EngineeringGhent UniversityCoupure Links 6539000GhentBelgium
| | - Matthias D'hooghe
- SynBioC Research GroupDepartment of Green Chemistry and TechnologyFaculty of Bioscience EngineeringGhent UniversityCoupure Links 6539000GhentBelgium
| |
Collapse
|
45
|
Huang YF, Zhu DJ, Chen XW, Chen QK, Luo ZT, Liu CC, Wang GX, Zhang WJ, Liao NZ. Curcumin enhances the effects of irinotecan on colorectal cancer cells through the generation of reactive oxygen species and activation of the endoplasmic reticulum stress pathway. Oncotarget 2018; 8:40264-40275. [PMID: 28402965 PMCID: PMC5522238 DOI: 10.18632/oncotarget.16828] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 02/27/2017] [Indexed: 12/20/2022] Open
Abstract
Although initially effective against metastatic colorectal cancer (CRC), irinotecan-based chemotherapy leads to resistance and adverse toxicity. Curcumin is well known for its anti-cancer effects in many cancers, including CRC. Here, we describe reactive oxygen species (ROS) generation and endoplasmic reticulum (ER) stress as important mechanisms by which curcumin enhances irinotecan's effects on CRC cells. CRC cell lines were treated with curcumin and/or irinotecan for 24 h, and then evaluated using cell proliferation assays, cell apoptosis assays, cell cycle analysis, intracellular Ca2+ measurements, ROS measurements and immunoblotting for key ER stress-related proteins. We found that cell viability was inhibited and apoptosis was increased, accompanied by ROS generation and ER stress activation in CRC cells treated with curcumin alone or in combination with irinotecan. Blocking ROS production attenuated the expression of two markers of ER stress: binding of immunoglobulin protein (BIP) and CCAAT/enhancer-binding protein homologous protein (CHOP). Blocking CHOP expression using RNA interference also inhibited ROS generation. These results demonstrated that curcumin could enhance the effects of irinotecan on CRC cells by inhibiting cell viability and inducing cell cycle arrest and apoptosis, and that these effects may be mediated, in part, by ROS generation and activation of the ER stress pathway.
Collapse
Affiliation(s)
- Yan-Feng Huang
- Department of Traditional Chinese Medicine, Shunde Hospital of Southern Medical University, Guangdong 528300, China
| | - Da-Jian Zhu
- Department of Gastrointestinal Surgery, Shunde Women and Children's Hospital Affiliated to Jinan University, Guangdong 528300, China
| | - Xiao-Wu Chen
- Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, Guangdong 528300, China
| | - Qi-Kang Chen
- Department of Gastrointestinal Surgery, Shunde Women and Children's Hospital Affiliated to Jinan University, Guangdong 528300, China
| | - Zhen-Tao Luo
- Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, Guangdong 528300, China
| | - Chang-Chun Liu
- Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, Guangdong 528300, China
| | - Guo-Xin Wang
- Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, Guangdong 528300, China
| | - Wei-Jie Zhang
- Department of Gastrointestinal Surgery, Shunde Hospital of Southern Medical University, Guangdong 528300, China
| | - Nv-Zhu Liao
- Department of Gastrointestinal Surgery, Shunde Women and Children's Hospital Affiliated to Jinan University, Guangdong 528300, China
| |
Collapse
|
46
|
Shubin AV, Demidyuk IV, Komissarov AA, Rafieva LM, Kostrov SV. Cytoplasmic vacuolization in cell death and survival. Oncotarget 2018; 7:55863-55889. [PMID: 27331412 PMCID: PMC5342458 DOI: 10.18632/oncotarget.10150] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 06/06/2016] [Indexed: 12/15/2022] Open
Abstract
Cytoplasmic vacuolization (also called cytoplasmic vacuolation) is a well-known morphological phenomenon observed in mammalian cells after exposure to bacterial or viral pathogens as well as to various natural and artificial low-molecular-weight compounds. Vacuolization often accompanies cell death; however, its role in cell death processes remains unclear. This can be attributed to studying vacuolization at the level of morphology for many years. At the same time, new data on the molecular mechanisms of the vacuole formation and structure have become available. In addition, numerous examples of the association between vacuolization and previously unknown cell death types have been reported. Here, we review these data to make a deeper insight into the role of cytoplasmic vacuolization in cell death and survival.
Collapse
Affiliation(s)
- Andrey V Shubin
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia.,Laboratory of Chemical Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia.,Laboratory of Biologically Active Nanostructures, N.F. Gamaleya Institute of Epidemiology and Microbiology, Moscow, Russia
| | - Ilya V Demidyuk
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia
| | - Alexey A Komissarov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia
| | - Lola M Rafieva
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia
| | - Sergey V Kostrov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Moscow, Russia
| |
Collapse
|
47
|
Wang J, Li XM, Bai Z, Chi BX, Wei Y, Chen X. Curcumol induces cell cycle arrest in colon cancer cells via reactive oxygen species and Akt/ GSK3β/cyclin D1 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2018; 210:1-9. [PMID: 28684297 DOI: 10.1016/j.jep.2017.06.037] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 02/10/2017] [Accepted: 06/10/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcuma kwangsiensis S. G. Lee & C. F. Liang (Guangxi ezhu, in Chinese) belongs to the Zingiberaceae family, has been used as a traditionally Chinese medicine nearly 2000 year. Curcumol is one of the guaiane-type sesquiterpenoid hemiketal isolated from medicine plant Curcuma kwangsiensis S. G. Lee & C. F. Liang, which has been reported possesses anti-cancer effects. Our previous study found that the most contribution to inhibit nasopharyngeal carcinoma cell growth was curcumol. AIM OF THE STUDY To assess the effect of curcumol on cell cycle arrest against human colon cancer cells (CRC) cells (LoVo and SW480) and explore its mechanism in vitro and in vivo. MATERIALS AND METHODS Curcumol was dissolved in absolute ethyl alcohol. The concentration of absolute ethyl alcohol in the control group or in experimental samples was always 1/500 (v/v) of the final medium volume. LoVo and SW480 cells were treated with different concentrations of curcumol (0, 53, 106, 212 and 424μM). And then the cell cycle of each group was examined by flow cytometry. The protein levels of PI3K, p-Akt, cyclin D1, cyclin E, CDK2, CDK4 and GSK3β were determined by Western blot. The mRNA expression of PI3K, Akt, cyclin D1, CDK4, P27, p21, and P16 in the treated cells were analyzed by real-time RT-PCR. In addition, the antitumor activity of curcumol was evaluated in nude mice bearing orthotopic tumor implants. RESULTS Curcumol induced cell cycle arrest in G1/S phase. RT-qPCR and Western blot data showed that curcumol enhanced the expression of GSK3β, P27, p21 and P16, and decreased the levels of PI3K, phosphorylated Akt (p-Akt), cyclin D1, CDK4, cyclin E and CDK2. Furthermore, curcumol induced reactive oxygen species (ROS) generation in LoVo cells, and ROS scavenger N-acetylcysteine (NAC) significantly reversed curcumol-induced cell growth inhibition. Besides, curcumol also prevented the growth of human colon cancer cells xenografts in nude mouse, accompanied by the reduction of PI3K, Akt, cyclin D1, CDK4, cycln E and significant increase of GSK3β. CONCLUSIONS Curcumol caused cell cycle arrest at the G0/G1 phase by ROS production and Akt/ GSK3β/cyclin D1 pathways inactivation, indicating the potential of curcumol in the prevention of colon cancer carcinogenesis.
Collapse
Affiliation(s)
- Juan Wang
- Xiangya Hospital Central South University, Chang sha 410008, China; College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Xu-Mei Li
- College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Zhun Bai
- Intensive Care Unit, Zhuzhou Central Hospital, Zhuzhou 412007, China
| | - Bi-Xia Chi
- Digestive System Department, The Frist People's Hospital of YueYang, Yueyang 414000, China
| | - Yan Wei
- College of Pharmacy, Guilin Medical University, Guilin 541004, China
| | - Xu Chen
- College of Pharmacy, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|
48
|
Martínez-Torres AC, Reyes-Ruiz A, Benítez-Londoño M, Franco-Molina MA, Rodríguez-Padilla C. IMMUNEPOTENT CRP induces cell cycle arrest and caspase-independent regulated cell death in HeLa cells through reactive oxygen species production. BMC Cancer 2018; 18:13. [PMID: 29298674 PMCID: PMC5753472 DOI: 10.1186/s12885-017-3954-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 12/21/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Regulated cell death (RCD) is a mechanism by which the cell activates its own machinery to self-destruct. RCD is important for the maintenance of tissue homeostasis and its deregulation is involved in diseases such as cervical cancer. IMMUNEPOTENT CRP (I-CRP) is a dialyzable bovine leukocyte extract that contains transfer factors and acts as an immunomodulator, and can be cytotoxic to cancer cell lines and reduce tumor burden in vivo. Although I-CRP has shown to improve or modulate immune response in inflammation, infectious diseases and cancer, its widespread use has been limited by the absence of conclusive data on the molecular mechanism of its action. METHODS In this study we analyzed the mechanism by which I-CRP induces cytotoxicity in HeLa cells. We assessed cell viability, cell death, cell cycle, nuclear morphology and DNA integrity, caspase dependence and activity, mitochondrial membrane potential, and reactive oxygen species production. RESULTS I-CRP diminishes cell viability in HeLa cells through a RCD pathway and induces cell cycle arrest in the G2/M phase. We show that the I-CRP induces caspase activation but cell death induction is independent of caspases, as observed by the use of a pan-caspase inhibitor, which blocked caspase activity but not cell death. Moreover, we show that I-CRP induces DNA alterations, loss of mitochondrial membrane potential, and production of reactive-oxygen species. Finally, pretreatment with N-acetyl-L-cysteine (NAC), a ROS scavenger, prevented both ROS generation and cell death induced by I-CRP. CONCLUSIONS Our data indicate that I-CRP treatment induced cell cycle arrest in G2/M phase, mitochondrial damage, and ROS-mediated caspase-independent cell death in HeLa cells. This work opens the way to the elucidation of a more detailed cell death pathway that could potentially work in conjunction with caspase-dependent cell death induced by classical chemotherapies.
Collapse
Affiliation(s)
- Ana Carolina Martínez-Torres
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología, 66455 Monterrey, Mexico
| | - Alejandra Reyes-Ruiz
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología, 66455 Monterrey, Mexico
| | - Milena Benítez-Londoño
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología, 66455 Monterrey, Mexico
| | - Moises Armides Franco-Molina
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología, 66455 Monterrey, Mexico
| | - Cristina Rodríguez-Padilla
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología, 66455 Monterrey, Mexico
| |
Collapse
|
49
|
Chen S, Nimick M, Cridge AG, Hawkins BC, Rosengren RJ. Anticancer potential of novel curcumin analogs towards castrate-resistant prostate cancer. Int J Oncol 2017; 52:579-588. [PMID: 29207190 DOI: 10.3892/ijo.2017.4207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/14/2017] [Indexed: 11/05/2022] Open
Abstract
Prostate cancer is initially sensitive to hormone therapy; however, over time the majority of patients progress to a hormone-insensitive form classified as castration-resistant prostate cancer (CRPC). CRPC is highly metastatic and patients have a poor prognosis. Thus, new drugs for the treatment of this disease are required. In this study, we therefore examined the cytotoxic effects and anticancer mechanism(s) of action of second generation curcumin analogs towards CRPC cells. For this purpose, PC3 and DU145 cells were treated with a series of curcumin analogs at 0-10 µM for 72 h and cytotoxicity was determined by the sulforhodamine B (SRB) assay. Two compounds, 1-isopropyl-3,5-bis(pyridin-3-ylmethylene)-4-piperidone (RL118) and 1-methyl-3,5-[(6-methoxynaphthalen-2-yl)methylene]-4-piperidone (RL121), were found to have the most potent cytotoxic effect with EC50 values of 0.50 and 0.58 µM in the PC3 cells and EC50 values of 0.76 and 0.69 µM in the DU145 cells, respectively. Thus, further experiments were performed focusing on these two compounds. Flow cytometry was performed to determine their effects on the cell cycle and apoptosis. Both analogs increased the number of cells in the G2/M phase of the cell cycle and induced apoptosis. Specifically, in the PC3 cells, RL121 increased the number of cells in the G2/M phase by 86% compared to the control, while RL118 increased the number of cells in the G2/M phase by 42% compared to the control after 24 h. Moreover, both RL118 and RL121 induced the apoptosis of both cell lines. In the DU145 cells, a 38-fold increase in the number of apoptotic cells was elicited by RL118 and a 78-fold increase by RL121 compared to the control. Furthermore, the effects of both analogs on the expression of key proteins involved in cell proliferation were also determined by western blot analysis. The results revealed that both analogs inhibited the expression of nuclear factor (NF)-κB (p65/RelA), eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), p-4E-BP1, mammalian target of rapamycin (mTOR), p-mTOR, AKT and p-AKT. Thus, the findings of this study provide evidence that RL118 and RL121 have potent anticancer activity against CPRC cells, and both analogs warrant further investigation in vivo.
Collapse
Affiliation(s)
- Shuli Chen
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Mhairi Nimick
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Andrew G Cridge
- Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand
| | - Bill C Hawkins
- Department of Chemistry, University of Otago, Dunedin 9016, New Zealand
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
50
|
Ying HZ, Chen Q, Zhang WY, Zhang HH, Ma Y, Zhang SZ, Fang J, Yu CH. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (Review). Mol Med Rep 2017; 16:7879-7889. [PMID: 28983598 PMCID: PMC5779870 DOI: 10.3892/mmr.2017.7641] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
The platelet‑derived growth factor (PDFG) signaling pathway exerts persistent activation in response to a variety of stimuli and facilitates the progression of hepatic fibrosis. Since this pathway modulates a broad spectrum of cellular processes, including cell growth, differentiation, inflammation and carcinogenesis, it has emerged as a therapeutic target for hepatic fibrosis and liver‑associated disorders. The present review exhibits the current knowledge of the role of the PDGF signaling pathway and its pathological profiles in hepatic fibrosis, and assesses the potential of inhibitors which have been investigated in the experimental hepatic fibrosis model, in addition to the clinical challenges associated with these inhibitors.
Collapse
Affiliation(s)
- Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Qin Chen
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Wen-You Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Huan-Huan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Yue Ma
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Song-Zhao Zhang
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jie Fang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|