1
|
Shu R, Yu Z, Wu J, Cheng Q, Peng Z, Zhou H, Zhao M. Inhibition of id-1 reduces osteosarcoma growth and metastasis through mediation of snail. J Orthop Surg Res 2025; 20:124. [PMID: 39891119 PMCID: PMC11784002 DOI: 10.1186/s13018-024-05412-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/28/2024] [Accepted: 12/24/2024] [Indexed: 02/03/2025] Open
Abstract
OBJECTIVE Osteosarcoma (OS) is a highly invasive bone tumor that frequently metastasizes to the lungs. This study aims to investigate the role of the Id-1 gene in OS invasion and metastasis, and its relationship with the Snail gene. METHODS This study included tissue samples from 12 non-metastatic osteosarcomas and 9 metastatic osteosarcoma patients to examine the expression of Id-1 and Snail using RT-qPCR and analyze their correlation. In cell-based experiments, four osteosarcoma cell lines (Saos-2, U2OS, MG-63, and 143B) and the human osteoblast cell line hFOB 1.19 were cultured. The expression of Id-1 and Snail was evaluated by RT-qPCR and Western blotting.Cells were randomly divided into the Control group, sh-NC group, and sh-Id-1 group using lentiviral infection. Transwell invasion and scratch assays were used to assess cell migration and invasion. WB was employed to detect the expression of Id-1, Snail, and epithelial-mesenchymal transition (EMT)-related proteins (E-cadherin, vimentin, and N-cadherin) in the OS cells of each group. In animal experiments, Tumor formation in each group was evaluated by injecting cells subcutaneously into mice. An osteosarcoma lung metastasis model was established by injecting infected cells into the tibia of mice. Tumor growth and lung metastasis were observed using HE staining. The expression of Id-1, Snail, and EMT-related proteins in osteosarcoma and lung tissues from each group of mice was assessed using Western blot and immunohistochemistry. RESULTS The expression of Id-1 and Snail was significantly higher in osteosarcoma tissues than in normal bone tissues, and the expression of Id-1 was positively correlated with that of Snail. In cell experiments, downregulation of Id-1 reduced Snail expression and significantly inhibited EMT, as well as the migration and invasion of OS cells (P < 0.05). In animal experiments, compared to the Control group, the sh-Id-1 group mice was no significant change in body weight, but the tumor volume was significantly reduced, and fewer lung metastatic nodules (P < 0.05). HE staining indicated decreased nuclear atypia, reduced invasion and destruction, fewer new blood vessels, and less calcification in the sh-Id-1 group tumors. Immunohistochemistry and WB results showed upregulation of E-cadherin and downregulation of vimentin, N-cadherin, Id-1, and Snail in the sh-Id-1 group (P < 0.05). CONCLUSION Downregulation of Id-1 inhibits the EMT process by reducing Snail expression, effectively suppressing the growth, invasion, and lung metastasis of OS.
Collapse
Affiliation(s)
- Rongbing Shu
- Department of Orthopedic, Yingtan People's Hospital, Yingtan, 335000, Jiangxi, China
| | - Zhuanyi Yu
- Department of Orthopedic, Yingtan People's Hospital, Yingtan, 335000, Jiangxi, China
| | - Jianmin Wu
- Department of Orthopedic, Yingtan People's Hospital, Yingtan, 335000, Jiangxi, China
| | - Qiuxin Cheng
- Department of Orthopedic, Yingtan People's Hospital, Yingtan, 335000, Jiangxi, China
| | - Zhihao Peng
- Department of Orthopedic, Yingtan People's Hospital, Yingtan, 335000, Jiangxi, China
| | - Huaqiang Zhou
- Department of Orthopedic, Yingtan People's Hospital, Yingtan, 335000, Jiangxi, China
| | - Min Zhao
- Department of Orthopedic, Yingtan People's Hospital, Yingtan, 335000, Jiangxi, China.
| |
Collapse
|
2
|
Stępniak J, Karbownik-Lewińska M. Protective Effects of Melatonin against Carcinogen-Induced Oxidative Damage in the Thyroid. Cancers (Basel) 2024; 16:1646. [PMID: 38730600 PMCID: PMC11083294 DOI: 10.3390/cancers16091646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/26/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Melatonin, primarily synthesized in the pineal gland, plays a crucial role in regulating circadian rhythms and possesses significant antioxidative properties. By neutralizing free radicals and reducing oxidative stress, melatonin emerges as a promising agent for the prevention and therapy of many different disorders, including cancer. This paper reviews the relationship between the thyroid gland and melatonin, presenting experimental evidence on the protective effects of this indoleamine against oxidative damage to macromolecules in thyroid tissue caused by documented carcinogens (as classified by the International Agency for Research on Cancer, IARC) or caused by potential carcinogens. Furthermore, the possible influence on cancer therapy in humans and the overall well-being of cancer patients are discussed. The article highlights melatonin's essential role in maintaining thyroid health and its contribution to management strategies in patients with thyroid cancer and other thyroid diseases.
Collapse
Affiliation(s)
- Jan Stępniak
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, Rzgowska St. 281/289, 93-338 Lodz, Poland;
| | - Małgorzata Karbownik-Lewińska
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, Rzgowska St. 281/289, 93-338 Lodz, Poland;
- Polish Mother’s Memorial Hospital-Research Institute, Rzgowska St. 281/289, 93-338 Lodz, Poland
| |
Collapse
|
3
|
Martínez-Campa C, Álvarez-García V, Alonso-González C, González A, Cos S. Melatonin and Its Role in the Epithelial-to-Mesenchymal Transition (EMT) in Cancer. Cancers (Basel) 2024; 16:956. [PMID: 38473317 DOI: 10.3390/cancers16050956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2024] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) is a cell-biological program that occurs during the progression of several physiological processes and that can also take place during pathological situations such as carcinogenesis. The EMT program consists of the sequential activation of a number of intracellular signaling pathways aimed at driving epithelial cells toward the acquisition of a series of intermediate phenotypic states arrayed along the epithelial-mesenchymal axis. These phenotypic features include changes in the motility, conformation, polarity and functionality of cancer cells, ultimately leading cells to stemness, increased invasiveness, chemo- and radioresistance and the formation of cancer metastasis. Amongst the different existing types of the EMT, type 3 is directly involved in carcinogenesis. A type 3 EMT occurs in neoplastic cells that have previously acquired genetic and epigenetic alterations, specifically affecting genes involved in promoting clonal outgrowth and invasion. Markers such as E-cadherin; N-cadherin; vimentin; and transcription factors (TFs) like Twist, Snail and ZEB are considered key molecules in the transition. The EMT process is also regulated by microRNA expression. Many miRNAs have been reported to repress EMT-TFs. Thus, Snail 1 is repressed by miR-29, miR-30a and miR-34a; miR-200b downregulates Slug; and ZEB1 and ZEB2 are repressed by miR-200 and miR-205, respectively. Occasionally, some microRNA target genes act downstream of the EMT master TFs; thus, Twist1 upregulates the levels of miR-10b. Melatonin is an endogenously produced hormone released mainly by the pineal gland. It is widely accepted that melatonin exerts oncostatic actions in a large variety of tumors, inhibiting the initiation, progression and invasion phases of tumorigenesis. The molecular mechanisms underlying these inhibitory actions are complex and involve a great number of processes. In this review, we will focus our attention on the ability of melatonin to regulate some key EMT-related markers, transcription factors and micro-RNAs, summarizing the multiple ways by which this hormone can regulate the EMT. Since melatonin has no known toxic side effects and is also known to help overcome drug resistance, it is a good candidate to be considered as an adjuvant drug to conventional cancer therapies.
Collapse
Affiliation(s)
- Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Virginia Álvarez-García
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| |
Collapse
|
4
|
de Morais JMB, Cruz EMS, Concato VM, de Souza MC, Santos YM, Quadreli DH, Inoue FSR, Ferreira FB, Fernandes GSA, Bidóia DL, Machado RRB, Chuffa LGA, Pavanelli WR, Seiva FRF. Unraveling the impact of melatonin treatment: Oxidative stress, metabolic responses, and morphological changes in HuH7.5 hepatocellular carcinoma cells. Pathol Res Pract 2024; 253:155056. [PMID: 38183817 DOI: 10.1016/j.prp.2023.155056] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/27/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/08/2024]
Abstract
In addition to its highly aggressive nature and late diagnosis, hepatocellular carcinoma (HCC) does not respond effectively to available chemotherapeutic agents. The search is on for an ideal and effective compound with low cost and minimal side effects that can be used as an adjunct to chemotherapeutic regimens. One of the mechanisms involved in the pathology of HCC is the oxidative stress, which plays a critical role in tumor survival and dissemination. Our group has already demonstrated the antitumor potential of melatonin against HuH 7.5 cells. In the present study, we focused on the effects of melatonin on oxidative stress parameters and their consequences on cell metabolism. HuH 7.5 cells were treated with 2 and 4 mM of melatonin for 24 and 48 h. Oxidative stress biomarkers, antioxidant enzyme, mitochondrial membrane potential, formation of lipid bodies and autophagic vacuoles, cell cycle progression, cell death rate and ultrastructural cell alterations were evaluated. The treatment with melatonin increased oxidative stress biomarkers and reduced antioxidant enzyme activities of HuH 7.5 cells. Additionally, melatonin treatment damaged the mitochondrial membrane and increased lipid bodies and autophagic vacuole formation. Melatonin triggered cell cycle arrest and induced cell death by apoptosis. Our results indicate that the treatment of HuH 7.5 cells with melatonin impaired antioxidant defense systems, inhibited cell cycle progression, and caused metabolic stress, culminating in tumor cell death.
Collapse
Affiliation(s)
- Juliana M B de Morais
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, State University of Londrina (UEL), PR, Brazil
| | - Ellen M S Cruz
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, State University of Londrina (UEL), PR, Brazil
| | - Virgínia M Concato
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, State University of Londrina (UEL), PR, Brazil
| | - Milena C de Souza
- North of Paraná State University (UENP), Biological Science Center, Bandeirantes, PR, Brazil
| | - Yasmin M Santos
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, State University of Londrina (UEL), PR, Brazil
| | - Débora H Quadreli
- General Biology Department, Biological Sciences Center, State University of Londrina, Londrina (UEL), PR, Brazil
| | - Fabrício S R Inoue
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, State University of Londrina (UEL), PR, Brazil
| | - Francielle B Ferreira
- North of Paraná State University (UENP), Biological Science Center, Bandeirantes, PR, Brazil
| | - Glaura S A Fernandes
- General Biology Department, Biological Sciences Center, State University of Londrina, Londrina (UEL), PR, Brazil
| | | | | | - Luiz Gustavo A Chuffa
- Department of Structural and Functional Biology, São Paulo State University (UNESP), Institute of Bioscience, Botucatu, SP, Brazil
| | - Wander R Pavanelli
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, State University of Londrina (UEL), PR, Brazil
| | - Fábio R F Seiva
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu, SP, Brazil.
| |
Collapse
|
5
|
Yi S, Luo M, Peng Y, Chen Y, Yu D. Anti-oncogenic mechanism of KLF17 in colon cancer by repressing cell migration and invasion via FHL1 upregulation. CHINESE J PHYSIOL 2023; 66:534-545. [PMID: 38149566 DOI: 10.4103/cjop.cjop-d-23-00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/28/2023] Open
Abstract
Colon cancer is a disease with high prevalence worldwide. This study sought to investigate Kruppel-like factor 17 (KLF17) mechanism in the development of colon cancer through four-and-a-half-LIM domain protein 1 (FHL1). In colon cancer cells, KLF17 and FHL1 expression was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot. After gain- and loss-of-function experiments in colon cancer cells, cell proliferative, invasive, and migrating abilities were tested by cell counting kit-8, transwell, and scratch assays, respectively. The expression of epithelial-mesenchymal transition (EMT)-related genes, E-cadherin, N-cadherin, and Vimentin, was measured by RT-qPCR and Western blot. Chromatin immunoprecipitation and dual-luciferase reporter gene assays were performed to detect the binding of KLF17 and the FHL1 promoter. Finally, a transplantation tumor model in nude mice was established for in vivo validation. Mechanistically, KLF17 facilitated FHL1 transcription by binding to the FHL1 promoter. KLF17 or FHL1 upregulation suppressed the colon cancer cell proliferative, invasive, and migrating capacities, accompanied by elevated E-cadherin expression and diminished N-cadherin and Vimentin expression. Furthermore, FHL1 silencing abrogated the repressive impacts of KLF17 upregulation on colon cancer cell EMT, proliferative, invasive, and migrating capabilities. Furthermore, KLF17 augmented FHL1 expression and curtailed the growth of transplanted tumors in nude mice. Conclusively, KLF17 promoted FHL1 transcription, thereby impeding the invasion, migration, and EMT of colon cancer cells.
Collapse
Affiliation(s)
- Shengen Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Luo
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanjin Peng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Chen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dan Yu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Polvat T, Prasertporn T, Na Nakorn P, Pannengpetch S, Suwanjang W, Panmanee J, Ngampramuan S, Cornish JL, Chetsawang B. Proteomic Analysis Reveals the Neurotoxic Effects of Chronic Methamphetamine Self-Administration-Induced Cognitive Impairments and the Role of Melatonin-Enhanced Restorative Process during Methamphetamine Withdrawal. J Proteome Res 2023; 22:3348-3359. [PMID: 37676068 PMCID: PMC10563163 DOI: 10.1021/acs.jproteome.3c00502] [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] [Academic Contribution Register] [Received: 08/09/2023] [Indexed: 09/08/2023]
Abstract
Cognitive flexibility is a crucial ability in humans that can be affected by chronic methamphetamine (METH) addiction. The present study aimed to elucidate the mechanisms underlying cognitive impairment in mice chronically administered METH via an oral self-administration method. Further, the effect of melatonin treatment on recovery of METH-induced cognitive impairment was also investigated. Cognitive performance of the mice was assessed using an attentional set shift task (ASST), and possible underlying neurotoxic mechanisms were investigated by proteomic and western blot analysis of the prefrontal cortex (PFC). The results showed that mice-administered METH for 21 consecutive days exhibited poor cognitive performance compared to controls. Cognitive deficit in mice partly recovered after METH withdrawal. In addition, mice treated with melatonin during METH withdrawal showed a higher cognitive recovery than vehicle-treated METH withdrawal mice. Proteomic and western blot analysis revealed that METH self-administration increased neurotoxic markers, including disruption to the regulation of mitochondrial function, mitophagy, and decreased synaptic plasticity. Treatment with melatonin during withdrawal restored METH-induced mitochondria and synaptic impairments. These findings suggest that METH-induced neurotoxicity partly depends on mitochondrial dysfunction leading to autophagy-dependent cell death and that the recovery of neurological impairments may be enhanced by melatonin treatment during the withdrawal period.
Collapse
Affiliation(s)
- Tanthai Polvat
- Research
Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
- Center
of Emotional Health, Department of Psychology, Macquarie University, Balaclava Road, North Ryde, NSW 2109, Australia
| | - Tanya Prasertporn
- Research
Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Piyada Na Nakorn
- Center
for Research Innovation and Bioinformatics, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Supitcha Pannengpetch
- Center
for Research Innovation and Bioinformatics, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Wilasinee Suwanjang
- Center
for Research Innovation and Bioinformatics, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Jiraporn Panmanee
- Research
Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Sukhonthar Ngampramuan
- Research
Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Jennifer L. Cornish
- Center
of Emotional Health, Department of Psychology, Macquarie University, Balaclava Road, North Ryde, NSW 2109, Australia
| | - Banthit Chetsawang
- Research
Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| |
Collapse
|
7
|
Yang X, Tian X, Zhao P, Wang Z, Sun X. Paclitaxel inhibits hepatocellular carcinoma tumorigenesis by regulating the circ_0005785/miR-640/GSK3β. Cell Biol Int 2023. [PMID: 37269228 DOI: 10.1002/cbin.11906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/03/2021] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 06/05/2023]
Abstract
Paclitaxel (PTX) is an effective chemotherapeutic agent for cancer patients. It has been reported that circular RNA (circRNA) circ_0005785is involved in the progression of hepatocellular carcinoma (HCC). The purpose of this study is to explore the role and mechanism of circ_0005785 in the PTX resistance of HCC. Cell viability, proliferation, invasion, migration, apoptosis, and angiogenesis were detected using 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), colony formation, transwell, wound-healing, flow cytometry, and tube formation assay. Circ_0005785, microRNA-640 (miR-640), and Glycogen synthase kinase-3 beta (GSK3β) levels were detected using real-time quantitative polymerase chain reaction. Protein levels of Proliferating cell nuclear antigen (PCNA), Bcl-2, and GSK3β were measured using western blot assay. After being predicted using Circular RNA interactome or TargetScan, binding between miR-640 and circ_0005785 or GSK3β was verified using dual-luciferase reporter and RNA Immunoprecipitation assay. PTX treatment could repress HCC cell viability, decrease circ_0005785 and GSK3β expression, and increase the miR-640 level in HCC cell lines. Furthermore, circ_0005785 and GSK3β were increased, and miR-640 was decreased in HCC tissues and cell lines. Moreover, circ_0005785 knockdown hindered proliferation, migration, invasion, angiogenesis, and boosted apoptosis in PTX-treated HCC cells in vitro. In addition, circ_0005785 silencing improved the PTX sensitivity of HCC in vivo. Mechanistically, circ_0005785 acted as a sponge of miR-640 to regulate GSK3β expression. PTX restrained HCC tumorigenesis partly via regulating the circ_0005785/miR-640/GSK3β axis, hinting at a promising therapeutic target for the HCC treatment.
Collapse
Affiliation(s)
- Xianwu Yang
- Department of Gastroenterology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xiaojuan Tian
- Department of Gastroenterology, Shenzhen University General Hospital, Shenzhen, China
| | - Pengcheng Zhao
- Department of Gastroenterology, Chengdu Seventh People's Hospital, Chengdu, China
| | - Zheng Wang
- Hepatobiliary Surgery, Huai'an Second People's Hospital/Huai'an Hospital, Xuzhou Medical University, Jiangsu, China
| | - Xuedong Sun
- Department of Gastroenterology, Shijiazhuang People's Hospital, Shijiazhuang, China
| |
Collapse
|
8
|
Estirado S, Fernández-Delgado E, Viñuelas-Zahínos E, Luna-Giles F, Rodríguez AB, Pariente JA, Espino J. Pro-Apoptotic and Anti-Migration Properties of a Thiazoline-Containing Platinum(II) Complex in MDA-MB-231 Breast Cancer Cells: The Role of Melatonin as a Synergistic Agent. Antioxidants (Basel) 2022; 11:1971. [PMID: 36290694 PMCID: PMC9598564 DOI: 10.3390/antiox11101971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/01/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/28/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive cancer insensitive to hormonal and human epidermal growth factor receptor 2 (HER2)-targeted therapies and has a poor prognosis. Therefore, there is a need for the development of convenient anticancer strategies for the management of TNBC. In this paper, we evaluate the antitumoral potential of a platinum(II) complex coordinated with the ligand 2-(3,5-diphenylpyrazol-1-yl)-2-thiazoline (DPhPzTn), hereafter PtDPhPzTn, against the TNBC cell line MDA-MB-231, and compared its effect with both cisplatin and its less lipophilic counterpart PtPzTn, the latter containing the ligand 2-(pyrazol-1-yl)-2-thiazoline (PzTn). Then, the putative potentiating actions of melatonin, a naturally occurring antioxidant with renowned antitumor properties, on the tumor-killing ability of PtDPhPzTn were also checked in TNBC cells. Our results show that PtDPhPzTn presented enhanced cytotoxicity compared to both the classical drug cisplatin and PtPzTn. In addition, PtDPhPzTn was able to induce apoptosis, being more selective for MDA-MB-231 cells when compared to non-tumor breast epithelial MCF10A cells. Likewise, PtDPhPzTn produced moderate S phase arrest and greatly impaired the migration ability of MDA-MB-231 cells. Most importantly, the co-stimulation of TNBC cells with PtDPhPzTn and melatonin substantially enhanced apoptosis and markedly improved the anti-migratory action compared to PtDPhPzTn alone. Altogether, our findings provide evidence that PtDPhPzTn and melatonin could be potentially applied to breast cancer treatment as powerful synergistic agents.
Collapse
Affiliation(s)
- Samuel Estirado
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Faculty of Science, University of Extremadura, 06006 Badajoz, Spain
| | - Elena Fernández-Delgado
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Faculty of Science, University of Extremadura, 06006 Badajoz, Spain
| | - Emilio Viñuelas-Zahínos
- Coordination Chemistry Research Group, Department of Organic and Inorganic Chemistry, Faculty of Science, University of Extremadura, 06006 Badajoz, Spain
| | - Francisco Luna-Giles
- Coordination Chemistry Research Group, Department of Organic and Inorganic Chemistry, Faculty of Science, University of Extremadura, 06006 Badajoz, Spain
| | - Ana B. Rodríguez
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Faculty of Science, University of Extremadura, 06006 Badajoz, Spain
| | - José A. Pariente
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Faculty of Science, University of Extremadura, 06006 Badajoz, Spain
| | - Javier Espino
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Faculty of Science, University of Extremadura, 06006 Badajoz, Spain
| |
Collapse
|
9
|
Design and optimization of PEGylated silver nanoparticles for efficient delivery of doxorubicin to cancer cells. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022]
|
10
|
Kumbul YÇ, Nazıroğlu M. Paclitaxel Promotes Oxidative Stress-Mediated Human Laryngeal Squamous Tumor Cell Death through the Stimulation of Calcium and Zinc Signaling Pathways: No Synergic Action of Melatonin. Biol Trace Elem Res 2022; 200:2084-2098. [PMID: 35075596 DOI: 10.1007/s12011-022-03125-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 12/14/2022]
Abstract
The paclitaxel (PAX) and melatonin (MLT)-mediated mitochondria reactive free oxygen radical (miROS) generations via the influx of excessive Ca2+ and Zn2+ induce tumor cell death and apoptosis. However, a presence of resistance was demonstrated against the PAX treatment in the tumor cells. The stimulation of TRPM2 may increase the anticancer action of PAX after the treatment of MLT. We investigated the stimulating role of PAX with/without MLT on the excessive Ca2+ influx and miROS generation-mediated human laryngeal squamous cancer (Hep2) cell death through the stimulation of TRPM2. The Hep2 cells were divided into four groups as control, MLT (1 mM for 2 h), PAX (50 μM for 24 h), and PAX + MLT. In some experiments, we induced additional subgroups such as PAX+ACA and PAX+2APB. The stimulation of TRPM2 induced the increase of TRPM2 current densities, lipid peroxidation, cytosolic ROS, miROS, cytosolic Ca2+, and Zn2+ values in the Hep2 cells after the treatment of PAX, although their values were decreased by the treatment of MLT and TRPM2 antagonists (ACA and 2APB). In addition, the PAX induced apoptosis and cell death via upregulation of caspases and downregulation of antioxidant glutathione peroxidase and glutathione in the cells. The treatment of PAX increased protein band expression values of TRPM2, PARP-1, and caspase 3 and 9 in the Hep2. The increased expression, apoptotic, and cell death values were not affected by the treatment of MLT. In conclusion, PAX induced the increase of Hep2 cell death via upregulations of TRPM2 and Zn2+, although its downregulation via the treatment of MLT did not change the antitumor action of PAX.
Collapse
Affiliation(s)
- Yusuf Çağdaş Kumbul
- Department of Otorhinolaryngology, Faculty of Medicine, Suleyman Demirel University, TR-32260, Isparta, Turkey
| | - Mustafa Nazıroğlu
- Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, TR-32260, Isparta, Turkey.
- BSN Health, Analyses, Innovation, Consultancy, Organization, Agriculture, Industry and Trade Limited Company, Göller Bölgesi Teknokenti, TR-32260, Isparta, Turkey.
| |
Collapse
|
11
|
Sadoughi F, Dana PM, Homayoonfal M, Sharifi M, Asemi Z. Molecular basis of melatonin protective effects in metastasis: A novel target of melatonin. Biochimie 2022; 202:15-25. [DOI: 10.1016/j.biochi.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/02/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022]
|
12
|
Ammar OA, El-Missiry MA, Othman AI, Amer ME. Melatonin is a potential oncostatic agent to inhibit HepG2 cell proliferation through multiple pathways. Heliyon 2022; 8:e08837. [PMID: 35141433 PMCID: PMC8814902 DOI: 10.1016/j.heliyon.2022.e08837] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2021] [Revised: 12/22/2021] [Accepted: 01/24/2022] [Indexed: 11/25/2022] Open
Abstract
CONTEXT Chemotherapy is a cornerstone in the treatment of hepatocellular carcinoma (HCC). Melatonin is a pineal hormone that targets various cancers, however, its antitumor pathways are still not fully elucidated. OBJECTIVE This study investigated melatonin's antitumor molecular mechanisms to inhibit the proliferation of HepG2 cells. MATERIALS AND METHODS HepG2 Cells were classified into cells without treatment as a control group and cells treated with melatonin (5.4 mmol/L) for 48 h. Proliferating cell nuclear antigen (PCNA) and marker of proliferation Ki-67 were estimated using immunohistochemical analysis. Apoptosis and cell cycle were evaluated using flow cytometric analysis. Apoptotic markers were detected using RT-qPCR assay. Antioxidants and oxidative stress biomarkers were performed using a colorimetric assay. RESULTS Melatonin produced a remarkable steady decrease in the viability of HepG2 cells at a concentration range between 5-20 mmol/L. Melatonin suppressed cell proliferation in the G2/M phase of the cell cycle (34.97 ± 0.92%) and induced apoptosis (12.43 ± 0.73%) through up-regulating p21 and p53 that was confirmed by the reduction of PCNA and Ki-67 expressions. Additionally, melatonin repressed angiogenesis evidenced by the down-regulation of angiopoietin-2, vascular endothelial growth factor receptor-2 expressions (0.42-fold change), and the level of CD133. Moreover, melatonin augmented the oxidative stress manifested by a marked increase of 4-hydroxynonenal levels with a reduction of glutathione content and superoxide dismutase activity. DISCUSSION AND CONCLUSION Melatonin inhibits proliferation and angiogenesis and induced apoptosis and oxidative stress in HepG2 cells. These results indicate the oncostatic effectiveness of melatonin on liver cancer.
Collapse
Affiliation(s)
- Omar A. Ammar
- Basic Science Department, Delta University for Science and Technology, Gamasa, Egypt
| | | | - Azza I. Othman
- Zoology Department, Faculty of Science, Mansoura University, Egypt
| | - Maggie E. Amer
- Zoology Department, Faculty of Science, Mansoura University, Egypt
| |
Collapse
|
13
|
Briguglio G, Costa C, Teodoro M, Giambò F, Italia S, Fenga C. Women's health and night shift work: Potential targets for future strategies in breast cancer (Review). Biomed Rep 2021; 15:98. [PMID: 34667595 PMCID: PMC8517754 DOI: 10.3892/br.2021.1474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2021] [Accepted: 08/12/2021] [Indexed: 11/18/2022] Open
Abstract
Breast cancer is the leading cause of cancer-associated amongst women worldwide. Several studies have shown that individual, environmental and occupational factors can serve an important role in the onset of breast cancer; although the majority of studies have demonstrated this association, and several studies have investigated the biological pathways, it is impossible to describe with certainty the causal relationship that involve circadian rhythm disruption and melatonin dysregulation with the oncogenic processes. Over the years, due to the introduction of more effective screening tools, an increase in the incidence of breast cancer as well as a decrease in the age at diagnosis has been witnessed. Subsequently, an increasing number of individuals have obtained care at a younger age, which has meant that after surgery and chemotherapy, these workers have had to return to work. In light of these paradigmatic changes, the aim of the present review was to identify potential targets for future organisational strategies that should be adopted in the workplace by occupational physicians, both for prevention and for the return-to-work process of working women who have suffered from breast cancer.
Collapse
Affiliation(s)
- Giusi Briguglio
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Occupational Medicine Section, University of Messina, I-98125 Messina, Italy
| | - Chiara Costa
- Department of Clinical and Experimental Medicine, University of Messina, I-98125 Messina, Italy
| | - Michele Teodoro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Occupational Medicine Section, University of Messina, I-98125 Messina, Italy
| | - Federica Giambò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Occupational Medicine Section, University of Messina, I-98125 Messina, Italy
| | - Sebastiano Italia
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Occupational Medicine Section, University of Messina, I-98125 Messina, Italy
| | - Concettina Fenga
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Occupational Medicine Section, University of Messina, I-98125 Messina, Italy
| |
Collapse
|
14
|
Abstract
Melatonin is a hormone that has many body functions and, for several decades, its antioxidant potential has been increasingly talked about. There is a relationship between failure in melatonin production in the pineal gland, an insufficient supply of this hormone to the body, and the occurrence of free radical etiology diseases such as neurodegenerative diseases, cardiovascular diseases, diabetes, cancer and others. Despite the development of molecular biology, numerous in vitro and in vivo studies, the exact mechanism of melatonin antioxidant activity is still unknown. Nowadays, the use of melatonin supplementation is more and more common, not only to prevent insomnia, but also to slow down the aging process and provide protection against diseases. The aim of this study is to get acquainted with current reports on melatonin, antioxidative mechanisms and their importance in diseases of free radical etiology.
Collapse
|
15
|
Wang X, Su P, Kang Y, Xu C, Qiu J, Wu J, Sheng P, Huang D, Zhang Z. Combination of Melatonin and Zoledronic Acid Suppressed the Giant Cell Tumor of Bone in vitro and in vivo. Front Cell Dev Biol 2021; 9:690502. [PMID: 34447747 PMCID: PMC8382950 DOI: 10.3389/fcell.2021.690502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/03/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Melatonin (Mlt) confers potential antitumor effects in various types of cancer. However, to the best of our knowledge, the role of Mlt in the giant cell tumor of bone (GCTB) remains unknown. Moreover, further research is required to assess whether Mlt can enhance the therapeutic effect of zoledronic acid (Zol), a commonly used anti-GCTB drug. In this research, we investigated the effects of Mlt, Zol, and the combination of these two drugs on GCTB cells’ characteristics, including cell proliferation, apoptosis, osteogenic differentiation, migration, and invasion. The cell counting kit-8 (CCK-8) assay, colony formation assay, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay (TUNEL), alkaline phosphatase (ALP) staining, alizarin red staining (ARS), scratch wound healing assay, and transwell experiment were performed, respectively. Our results showed that Mlt could effectively inhibit the proliferation, migration, and invasion of GCTB cells, as well as promote the apoptosis and osteogenic differentiation of tumor cells. Of note, a stronger antitumor effect was observed when Mlt was combined with Zol treatment. This therapeutic effect might be achieved by inhibiting the activation of both the Hippo and NF-κB pathways. In conclusion, our study suggests that Mlt can be a new treatment for GCTB, which could further enhance the antitumor effect of Zol.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Peiqiang Su
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Kang
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Caixia Xu
- Research Centre for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jincheng Qiu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinna Wu
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Puyi Sheng
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dongsheng Huang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ziji Zhang
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
Ezzati M, Velaei K, Kheirjou R. Melatonin and its mechanism of action in the female reproductive system and related malignancies. Mol Cell Biochem 2021; 476:3177-3190. [PMID: 33864572 DOI: 10.1007/s11010-021-04151-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/03/2020] [Accepted: 04/01/2021] [Indexed: 12/14/2022]
Abstract
Melatonin (N-acetyl-5-methoxytryptamine), the main product of pineal gland in vertebrates, is well known for its multifunctional role which has great influences on the reproductive system. Recent studies documented that melatonin is a powerful free radical scavenger that affects the reproductive system function and female infertility by MT1 and MT2 receptors. Furthermore, cancer researches indicate the influence of melatonin on the modulation of tumor cell signaling pathways resulting in growth inhibitor of the both in vivo/in vitro models. Cancer adjuvant therapy can also benefit from melatonin through therapeutic impact and decreasing the side effects of radiation and chemotherapy. This article reviews the scientific evidence about the influence of melatonin and its mechanism of action on the fertility potential, physiological alteration, and anticancer efficacy, during experimental and clinical studies.
Collapse
Affiliation(s)
- Maryam Ezzati
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Immunology Research Center, Tabriz University of Medical Sciences, PO. Box: 51376563833, Tabriz, Iran.
| | - Kobra Velaei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raziyeh Kheirjou
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Mayo V, Bowles AC, Wubker LE, Ortiz I, Cordoves AM, Cote RJ, Correa D, Agarwal A. Human-derived osteoblast-like cells and pericyte-like cells induce distinct metastatic phenotypes in primary breast cancer cells. Exp Biol Med (Maywood) 2021; 246:971-985. [PMID: 33210551 PMCID: PMC8024509 DOI: 10.1177/1535370220971599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/09/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Approximately 70% of advanced breast cancer patients will develop bone metastases, which accounts for ∼90% of cancer-related mortality. Breast cancer circulating tumor cells (CTCs) establish metastatic tumors in the bone after a close interaction with local bone marrow cells including pericytes and osteoblasts, both related to resident mesenchymal stem/stromal cells (BM-MSCs) progenitors. In vitro recapitulation of the critical cellular players of the bone microenvironment and infiltrating CTCs could provide new insights into their cross-talk during the metastatic cascade, helping in the development of novel therapeutic strategies. Human BM-MSCs were isolated and fractionated according to CD146 presence. CD146+ cells were utilized as pericyte-like cells (PLCs) given the high expression of the marker in perivascular cells, while CD146- cells were induced into an osteogenic phenotype generating osteoblast-like cells (OLCs). Transwell migration assays were performed to establish whether primary breast cancer cells (3384T) were attracted to OLC. Furthermore, proliferation of 3384T breast cancer cells was assessed in the presence of PLC- and OLC-derived conditioned media. Additionally, conditioned media cultures as well as transwell co-cultures of each OLCs and PLCs were performed with 3384T breast cancer cells for gene expression interrogation assessing their induced transcriptional changes with an emphasis on metastatic potential. PLC as well as their conditioned media increased motility and invasion potential of 3384T breast cancer cells, while OLC induced a dormant phenotype, downregulating invasiveness markers related with migration and proliferation. Altogether, these results indicate that PLC distinctively drive 3384T cancer cells to an invasive and migratory phenotype, while OLC induce a quiescence state, thus recapitulating the different phases of the in vivo bone metastatic process. These data show that phenotypic responses from metastasizing cancer cells are influenced by neighboring cells at the bone metastatic niche during the establishment of secondary metastatic tumors.
Collapse
Affiliation(s)
- Vera Mayo
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
| | - Annie C Bowles
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Laura E Wubker
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
| | - Ismael Ortiz
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
| | - Albert M Cordoves
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
| | - Richard J Cote
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St Louis, MO 63110, USA
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33146, USA
- Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
18
|
Cheng G, Ma T, Deng Z, Gutiérrez-Gamboa G, Ge Q, Xu P, Zhang Q, Zhang J, Meng J, Reiter RJ, Fang Y, Sun X. Plant-derived melatonin from food: a gift of nature. Food Funct 2021; 12:2829-2849. [PMID: 33877242 DOI: 10.1039/d0fo03213a] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
Abstract
In recent years, people have become increasingly interested in bioactive ingredients from plants, especially antioxidant molecules such as melatonin, which are beneficial to human health. The purpose of this article is to provide new information on plant-derived foods with a high content of melatonin. We comprehensively summarize the content of melatonin in plant-derived foods and discuss the factors that influence melatonin levels to provide new ideas on enhancement. Additionally, we describe the biosynthetic pathway of melatonin and identify its major functions. Medicinal herbs are often rich in melatonin while many vegetables and fruits exhibit somewhat lower levels with wide variations among species. The genetic traits of plants, the phenological stage of the cultivar, the photoperiod, the level of stress to which the plants are exposed at the time of harvest, exposure to agrochemicals and determination methods are the main factors affecting the melatonin content. To date, standardization of uniform sampling times and the use of suitable pretreatments as well as determination methods have not been achieved. The results of the studies reviewed highlight the potentially important role of plant melatonin in influencing the progression of human diseases. Based on the health promotional aspects of melatonin, consuming foods containing higher concentrations of tryptophan and melatonin is suggested.
Collapse
Affiliation(s)
- Guo Cheng
- College of Enology, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Gurunathan S, Qasim M, Kang MH, Kim JH. Role and Therapeutic Potential of Melatonin in Various Type of Cancers. Onco Targets Ther 2021; 14:2019-2052. [PMID: 33776451 PMCID: PMC7987311 DOI: 10.2147/ott.s298512] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/26/2020] [Accepted: 03/02/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is a large group of diseases and the second leading cause of death worldwide. Lung, prostate, colorectal, stomach, and liver cancers are the most common types of cancer in men, whereas breast, colorectal, lung, cervical, and thyroid cancers are the most common among women. Presently, various treatment strategies, including surgical resection combined with chemotherapy, radiotherapy, nanotherapy, and immunotherapy, have been used as conventional treatments for patients with cancer. However, the clinical outcomes of advanced-stage disease remain relatively unfavorable owing to the emergence of chemoresistance, toxicity, and other undesired detrimental side effects. Therefore, new therapies to overcome these limitations are indispensable. Recently, there has been considerable evidence from experimental and clinical studies suggesting that melatonin can be used to prevent and treat cancer. Studies have confirmed that melatonin mitigates the pathogenesis of cancer by directly affecting carcinogenesis and indirectly disrupting the circadian cycle. Melatonin (MLT) is nontoxic and exhibits a range of beneficial effects against cancer via apoptotic, antiangiogenic, antiproliferative, and metastasis-inhibitory pathways. The combination of melatonin with conventional drugs improves the drug sensitivity of cancers, including solid and liquid tumors. In this manuscript, we will comprehensively review some of the cellular, animal, and human studies from the literature that provide evidence that melatonin has oncostatic and anticancer properties. Further, this comprehensive review compiles the available experimental and clinical data analyzing the history, epidemiology, risk factors, therapeutic effect, clinical significance, of melatonin alone or in combination with chemotherapeutic agents or radiotherapy, as well as the underlying molecular mechanisms of its anticancer effect against lung, breast, prostate, colorectal, skin, liver, cervical, and ovarian cancers. Nonetheless, in the interest of readership clarity and ease of reading, we have discussed the overall mechanism of the anticancer activity of melatonin against different types of cancer. We have ended this report with general conclusions and future perspectives.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Muhammad Qasim
- Center of Bioengineering and Nanomedicine, Department of Food Science, University of Otago, Dunedin, 9054, New Zealand
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Korea
| |
Collapse
|
20
|
Griñan-Lison C, Blaya-Cánovas JL, López-Tejada A, Ávalos-Moreno M, Navarro-Ocón A, Cara FE, González-González A, Lorente JA, Marchal JA, Granados-Principal S. Antioxidants for the Treatment of Breast Cancer: Are We There Yet? Antioxidants (Basel) 2021; 10:205. [PMID: 33572626 PMCID: PMC7911462 DOI: 10.3390/antiox10020205] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/30/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most frequent cancer and the leading cause of cancer death in women. Oxidative stress and the generation of reactive oxygen species (ROS) have been related to cancer progression. Compared to their normal counterparts, tumor cells show higher ROS levels and tight regulation of REDOX homeostasis to maintain a low degree of oxidative stress. Traditionally antioxidants have been extensively investigated to counteract breast carcinogenesis and tumor progression as chemopreventive agents; however, there is growing evidence indicating their potential as adjuvants for the treatment of breast cancer. Aimed to elucidate whether antioxidants could be a reality in the management of breast cancer patients, this review focuses on the latest investigations regarding the ambivalent role of antioxidants in the development of breast cancer, with special attention to the results derived from clinical trials, as well as their potential use as plausible agents in combination therapy and their power to ameliorate the side effects attributed to standard therapeutics. Data retrieved herein suggest that antioxidants play an important role in breast cancer prevention and the improvement of therapeutic efficacy; nevertheless, appropriate patient stratification based on "redoxidomics" or tumor subtype is mandatory in order to define the dosage for future standardized and personalized treatments of patients.
Collapse
Affiliation(s)
- Carmen Griñan-Lison
- Centre for Biomedical Research (CIBM), Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, 18100 Granada, Spain; (C.G.-L.); (J.A.M.)
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18100 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Jose L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Araceli López-Tejada
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Marta Ávalos-Moreno
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Alba Navarro-Ocón
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Francisca E. Cara
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Adrián González-González
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Jose A. Lorente
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
- Department of Legal Medicine, School of Medicine, University of Granada, 18016 Granada, Spain
| | - Juan A. Marchal
- Centre for Biomedical Research (CIBM), Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, 18100 Granada, Spain; (C.G.-L.); (J.A.M.)
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18100 Granada, Spain
- Department of Human Anatomy and Embryology, School of Medicine, University of Granada, 18016 Granada, Spain
| | - Sergio Granados-Principal
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18011 Granada, Spain
| |
Collapse
|
21
|
Jardin I, Diez-Bello R, Falcon D, Alvarado S, Regodon S, Salido GM, Smani T, Rosado JA. Melatonin downregulates TRPC6, impairing store-operated calcium entry in triple-negative breast cancer cells. J Biol Chem 2021; 296:100254. [PMID: 33380424 PMCID: PMC7948746 DOI: 10.1074/jbc.ra120.015769] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/25/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 12/15/2022] Open
Abstract
Melatonin has been reported to induce effective reduction in growth and development in a variety of tumors, including breast cancer. In triple-negative breast cancer (TNBC) cells, melatonin attenuates a variety of cancer features, such as tumor growth and apoptosis resistance, through a number of still poorly characterized mechanisms. One biological process that is important for TNBC cells is store-operated Ca2+ entry (SOCE), which is modulated by TRPC6 expression and function. We wondered whether melatonin might intersect with this pathway as part of its anticancer activity. We show that melatonin, in the nanomolar range, significantly attenuates TNBC MDA-MB-231 cell viability, proliferation, and migration in a time- and concentration-dependent manner, without having any effect on nontumoral breast epithelial MCF10A cells. Pretreatment with different concentrations of melatonin significantly reduced SOCE in MDA-MB-231 cells without altering Ca2+ release from the intracellular stores. By contrast, SOCE in MCF10A cells was unaffected by melatonin. In the TNBC MDA-MB-468 cell line, melatonin not only attenuated viability, migration, and SOCE, but also reduced TRPC6 expression in a time- and concentration-dependent manner, without altering expression or function of the Ca2+ channel Orai1. The expression of exogenous TRPC6 overcame the effect of melatonin on SOCE and cell proliferation, and silencing or inhibition of TRPC6 impaired the inhibitory effect of melatonin on SOCE. These findings indicate that TRPC6 downregulation might be involved in melatonin's inhibitory effects on Ca2+ influx and the maintenance of cancer hallmarks and point toward a novel antitumoral mechanism of melatonin in TNBC cells.
Collapse
Affiliation(s)
- Isaac Jardin
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain.
| | - Raquel Diez-Bello
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Debora Falcon
- Cardiovascular Physiopathology Group, Institute of Biomedicine of Sevilla, Sevilla, Spain
| | - Sandra Alvarado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Sergio Regodon
- Department of Animal Medicine, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysic, Institute of Biomedicine of Sevilla, Sevilla, Spain
| | - Juan A Rosado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| |
Collapse
|
22
|
Maroufi NF, Ashouri N, Mortezania Z, Ashoori Z, Vahedian V, Amirzadeh-Iranaq MT, Fattahi A, Kazemzadeh H, Bizzarri M, Akbarzadeh M, Nejabati HR, Faridvand Y, Rashidi MR, Nouri M. The potential therapeutic effects of melatonin on breast cancer: An invasion and metastasis inhibitor. Pathol Res Pract 2020; 216:153226. [PMID: 32987338 DOI: 10.1016/j.prp.2020.153226] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/09/2020] [Revised: 08/30/2020] [Accepted: 09/11/2020] [Indexed: 01/14/2023]
Abstract
Breast cancer is the most common cancer among women and its metastasis which generally observed at the last stage is the major cause of breast cancer-related death. Therefore, the agents that have the potential to prevent metastatic and invasive nature of breast cancer can open up new therapeutic strategies. Melatonin, a major hormone of pineal gland, is a powerful anti-cancer agent. There are growing evidence regarding the protective effect of melatonin against cancer invasion and metastasis. The anti-metastatic feature of melatonin accompanies with suppression of tumor proliferation, induction of tumor apoptosis, regulation of the cell cycle, modulating angiogenesis, impediment of invasion, and induction of cancer cells sensitivity to the chemotherapy agents. More recently, anti-metastatic effect of melatonin through affecting cancer stem cells and vascular mimicry has been identified. Thus, the aim of this review is to discuss the potential therapeutic effect of melatonin on breast cancer via modulating the cells invasion and metastasis.
Collapse
Affiliation(s)
- Nazila Fathi Maroufi
- Student Research Committee, Tabriz University of Medical Sciences, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Ashouri
- Department of Medicine, Faculty of Cellular and Molecular Biology, University of Shiraz, Iran
| | | | - Zahra Ashoori
- Department of Medical, Faculty of Medical, University of Shahid Beheshti, Iran
| | - Vahid Vahedian
- Department of Experimental Biomedicine, Dr. Vahid Vahedian Medical Diagnostic Laboratory Gorgan, Iran; Department of Medical Laboratory Sciences, Faculty of Medicine, Islamic Azad University (IAU), Sari, Iran
| | | | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Kazemzadeh
- Department of Biology, Faculty of Basic Sciences, Hakim Sabzevari University, Iran
| | - Mariano Bizzarri
- University La Sapienza, Department of Experimental Medicine, System Biology Group, Rome, Italy
| | - Maryam Akbarzadeh
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Faridvand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Ma S, Wang F, Zhang C, Wang X, Wang X, Yu Z. Cell metabolomics to study the function mechanism of Cyperus rotundus L. on triple-negative breast cancer cells. BMC Complement Med Ther 2020; 20:262. [PMID: 32843016 PMCID: PMC7449030 DOI: 10.1186/s12906-020-02981-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/31/2019] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a kind of malignant tumor with higher recurrence and metastasis rate. According to historical records, the dry rhizomes Cyperus rotundus L. could be ground into powder and mixed with ginger juice and wine for external application for breast cancer. We studied the effect of the ethanol extract of Cyperus rotundus L. (EECR) on TNBC cells and found its' apoptosis-inducing effect with a dose-relationship. But the function mechanism of EECR on TNBC is still mysterious. Hence, the present research aimed to detect its function mechanism at the small molecule level through ultra-high performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UPLC-Q-TOF-MS/MS) metabolomics. METHODS The CCK-8 assay and the Annexin V-FITC/PI assay were applied to test the effect of EECR on MDA-MB-231 cells and MDA-MB 468 cells at various concentrations of 0, 200, 400, and 600 μg/ml. UPLC-Q-TOF-MS/MS based metabolomics was used between the control group and the EECR treatment groups. Multivariate statistical analysis was used to visualize the apoptosis-inducing action of EECR and filtrate significantly changed metabolites. RESULTS The apoptosis-inducing action was confirmed and forty-nine significantly changed metabolites (VIP > 1, p < 0.05, and FC > 1.2 or FC < 0.8) were identified after the interference of EECR. The level of significant differential metabolites between control group, middle dose group, and high dose group were compared and found that which supported the apoptosis-inducing action with dose-dependence. CONCLUSION By means of metabolism, we have detected the mechanism of EECR inducing apoptosis of TNBC cells at the level of small molecule metabolites and found that EECR impacted the energy metabolism of TNBC cells. In addition, we concluded that EECR induced apoptosis by breaking the balance between ATP-production and ATP-consumption: arresting the pathways of Carbohydrate metabolism such as Central carbon metabolism in cancer, aerobic glycolysis, and Amino sugar and nucleotide sugar metabolism, whereas accelerating the pathways of ATP-consumption including Amino Acids metabolism, Fatty acid metabolism, Riboflavin metabolism and Purine metabolism. Although further study is still needed, EECR has great potential in the clinical treatment of TNBC with fewer toxic and side effects.
Collapse
Affiliation(s)
- Shuangshuang Ma
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 jiyan road, Jinan, 250017, Shandong, China
- Shandong Hongjitang Pharmaceutical Group Co.,Ltd., Jinan, 250000, China
| | - Fukai Wang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 jiyan road, Jinan, 250017, Shandong, China
| | - Caijuan Zhang
- School of life Science, Beijing University of Chinese Medicine, Northeast corner of intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing, 102488, China
| | - Xinzhao Wang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 jiyan road, Jinan, 250017, Shandong, China
| | - Xueyong Wang
- School of Chinese Materia Medical, Beijing University of Chinese Medicine, No.11 North 3rd Ring East Road, Chao-Yang District, Beijing, 100029, China.
| | - Zhiyong Yu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 jiyan road, Jinan, 250017, Shandong, China.
| |
Collapse
|
24
|
Zhu X, Luo C, Lin K, Bu F, Ye F, Huang C, Luo H, Huang J, Zhu Z. Overexpression of DJ-1 enhances colorectal cancer cell proliferation through the cyclin-D1/MDM2-p53 signaling pathway. Biosci Trends 2020; 14:83-95. [PMID: 32132307 DOI: 10.5582/bst.2019.01272] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/05/2022]
Abstract
Emerging evidence indicates that DJ-1 is highly expressed in different cancers. It modulates cancer progression, including cell proliferation, cell apoptosis, invasion, and metastasis. However, its role in colorectal cancer (CRC) remains poorly defined. The current study noted increased DJ-1 expression in CRC tumor tissue and found that its expression was closely related to clinical-pathological features. Similarly, DJ-1 increased in CRC cells (SW480, HT-29, Caco-2, LoVo, HCT116, and SW620), and especially in SW480 and HCT116 cells. Functional analyses indicated that overexpression of DJ-1 promoted CRC cell invasion, migration, and proliferation in vitro and in vivo. Mechanistic studies indicated that DJ-1 increased in CRC cell lines, activated specific protein cyclin-D1, and modulated the MDM2/p53 signaling pathway by regulating the levels of the downstream factors Bax, Caspase-3, and Bcl-2, which are related to the cell cycle and apoptosis. Conversely, knockdown of DJ-1 upregulated p53 expression by disrupting the interaction between p53 and MDM2 and inhibiting CRC cell proliferation, revealing the pro-oncogenic mechanism of DJ-1 in CRC. In conclusion, the current findings provide compelling evidence that DJ-1 might be a promoter of CRC cell invasion, proliferation, and migration via the cyclin-D1/MDM2-p53 signaling pathway. Findings also suggest its potential role as a postoperative adjuvant therapy for patients with CRC.
Collapse
Affiliation(s)
- Xiaojian Zhu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Chen Luo
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Kang Lin
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Fanqin Bu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Fan Ye
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Chao Huang
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Hongliang Luo
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Jun Huang
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Zhengming Zhu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
25
|
Yang SF, Chen YS, Chien HW, Wang K, Lin CL, Chiou HL, Lee CY, Chen PN, Hsieh YH. Melatonin attenuates epidermal growth factor-induced cathepsin S expression in ARPE-19 cells: Implications for proliferative vitreoretinopathy. J Pineal Res 2020; 68:e12615. [PMID: 31605630 DOI: 10.1111/jpi.12615] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/19/2019] [Revised: 09/24/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022]
Abstract
Abnormal proliferation and motility of retinal pigment epithelial cells leads to proliferative vitreoretinopathy (PVR). Melatonin is a known effective antitumour and anti-invasive agent, but whether it affects the formation and underlying mechanisms of PVR remains unclear. In this study, the results of the MTT assay, colony formation and propidium iodide (PI) staining with flow cytometry revealed that melatonin dose dependently inhibited epidermal growth factor (EGF)-induced proliferation of human ARPE-19 cells. Furthermore, melatonin reduced EGF-induced motility by suppressing cathepsin S (CTSS) expression. Pretreatment with ZFL (a CTSS inhibitor) or overexpression of CTSS (pCMV-CTSS) significantly inhibited EGF-induced cell motility when combined with melatonin. Epidermal growth factor induced the phosphorylation of AKT(S473)/mTOR (S2448) and transcription factor (c-Jun/Sp1) signaling pathways. Pretreatment of LY294002 (a PI3K inhibitor) or rapamycin (an mTOR inhibitor) markedly reduced EGF-induced motility and p-AKT/p-mTOR/c-Jun/Sp1 expression when combined with melatonin. Taken together, these data indicate that melatonin inhibited EGF-induced proliferation and motility of human ARPE-19 cells by activating the AKT/mTOR pathway, which is dependent on CTSS modulation of c-Jun/Sp1 signalling. Melatonin may be a promising therapeutic drug against PVR.
Collapse
Affiliation(s)
- Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yong-Syuan Chen
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiang-Wen Chien
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Departments of Ophthalmology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
- Department of Ophthalmology, Cathay General Hospital, Taipei, Taiwan
| | - Kai Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Departments of Ophthalmology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
- Department of Ophthalmology, Cathay General Hospital, Taipei, Taiwan
| | - Chia-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Hui-Ling Chiou
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Yi Lee
- Department of Ophthalmology, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Pei-Ni Chen
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Clinical laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
26
|
Zhang J, Li G, Feng L, Lu H, Wang X. Krüppel-like factors in breast cancer: Function, regulation and clinical relevance. Biomed Pharmacother 2019; 123:109778. [PMID: 31855735 DOI: 10.1016/j.biopha.2019.109778] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2019] [Revised: 11/27/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022] Open
Abstract
Breast cancer has accounted for the leading cause of cancer-related mortality among women worldwide. Although the progress in its diagnosis and treatment has come at a remarkable pace during the past several decades, there are still a wide array of problems regarding its progression, metastasis and treatment resistance that have not yet been fully clarified. Recently, an increasing number of studies have revealed that some members of Krüppel-like factors(KLFs) are significantly associated with cell proliferation, apoptosis, metastasis, cancer stem cell regulation and prognostic and predictive value for patients in breast cancer, indicating their promising prognostic and predictive potential for breast cancer survival and outcome. In this review, we will summarize our current knowledge of the functions, regulations and clinical relevance of KLFs in breast cancer.
Collapse
Affiliation(s)
- Jianping Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China; Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Guangliang Li
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lifeng Feng
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Haiqi Lu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China; Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China; Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| |
Collapse
|
27
|
Singh VK, Arora D, Ansari MI, Sharma PK. Phytochemicals based chemopreventive and chemotherapeutic strategies and modern technologies to overcome limitations for better clinical applications. Phytother Res 2019; 33:3064-3089. [DOI: 10.1002/ptr.6508] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/17/2019] [Revised: 07/26/2019] [Accepted: 08/23/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Vipendra Kumar Singh
- Environmental Carcinogenesis Laboratory, Food, Drug and Chemical Toxicology GroupCSIR‐Indian Institute of Toxicology Research Lucknow India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad‐ 201002 India
| | - Deepika Arora
- Environmental Carcinogenesis Laboratory, Food, Drug and Chemical Toxicology GroupCSIR‐Indian Institute of Toxicology Research Lucknow India
- Material and Measurement LaboratoryNational Institute of Standards and Technology Gaithersburg 20899 Maryland USA
| | - Mohammad Imran Ansari
- Environmental Carcinogenesis Laboratory, Food, Drug and Chemical Toxicology GroupCSIR‐Indian Institute of Toxicology Research Lucknow India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad‐ 201002 India
| | - Pradeep Kumar Sharma
- Environmental Carcinogenesis Laboratory, Food, Drug and Chemical Toxicology GroupCSIR‐Indian Institute of Toxicology Research Lucknow India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad‐ 201002 India
| |
Collapse
|
28
|
Amin AH, El-Missiry MA, Othman AI, Ali DA, Gouida MS, Ismail AH. Ameliorative effects of melatonin against solid Ehrlich carcinoma progression in female mice. J Pineal Res 2019; 67:e12585. [PMID: 31066091 DOI: 10.1111/jpi.12585] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/18/2019] [Revised: 04/24/2019] [Accepted: 05/02/2019] [Indexed: 12/27/2022]
Abstract
The current work estimated the antitumour efficacy of melatonin (MLT) on the growth of Ehrlich ascites carcinoma cells inoculated intramuscularly into the hind limbs of female BALB/c mice and to compare its effects with those of adriamycin (ADR). After solid tumours developed, the animals were divided into the three following groups: the tumour-bearing control, MLT-treated (20 mg/kg body weight) and ADR-treated (10 mg/kg body weight) groups. The results showed a significant reduction in the tumour masses of the treated animals in comparison with those of the control group. There were a significant decrease in the malondialdehyde level and a significant elevation of the glutathione concentration and the superoxide dismutase and catalase activities in the MLT and ADR groups. The current study indicated the increased expression levels of P53, caspase-3 and caspase-9 and the decreased expression levels of the rRNA and Bcl2. The MLT and ADR treatments resulted in histological changes, such as a marked degenerative area, the necrosis of neoplastic cells, the appearance of different forms of apoptotic cells and giant cells with condensed chromatin, and a deeply eosinophilic cytoplasm. The MLT and ADR treatments also significantly decreased the Ki-67 protein and vascular endothelial growth factor (VEGF) expression levels in the tumour masses. In conclusion, similar to ADR-treated tumour-bearing mice, MLT suppressed the growth and proliferation of tumour by inducing apoptosis and by inhibiting tumour vascularization. The current data recommend MLT as a safe natural chemotherapeutic adjuvant to overcome cancer progression after a clinical trial validates these results.
Collapse
Affiliation(s)
- Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Azza I Othman
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Doaa A Ali
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Mona S Gouida
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Ahmed H Ismail
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
- Biology Department, Faculty of Science, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|