1
|
Pourbarkhordar V, Rahmani S, Roohbakhsh A, Hayes AW, Karimi G. Melatonin effect on breast and ovarian cancers by targeting the PI3K/Akt/mTOR pathway. IUBMB Life 2024; 76:1035-1049. [PMID: 39212097 DOI: 10.1002/iub.2900] [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: 04/02/2024] [Accepted: 05/29/2024] [Indexed: 09/04/2024]
Abstract
Melatonin, the hormone of the pineal gland, possesses a range of physiological functions, and recently, its anticancer effect has become more apparent. A more thorough understanding of molecular alterations in the components of several signaling pathways as new targets for cancer therapy is needed because of current innate restrictions such as drug toxicity, side effects, and acquired or de novo resistance. The PI3K/Akt/mTOR pathway is overactivated in many solid tumors, such as breast and ovarian cancers. This pathway in normal cells is essential for growth, proliferation, and survival. However, it is an undesirable characteristic in malignant cells. We have reviewed multiple studies about the effect of melatonin on breast and ovarian cancer, focusing on the PI3K/Akt/mTOR pathway. Melatonin exerts its inhibitory effects via several mechanisms. A: Downregulation of downstream or upstream components of the signaling pathway such as phosphatase and tensin homolog (PTEN), phosphatidylinositol (3,4,5)-trisphosphate kinase (PI3K), p-PI3K, Akt, p-Akt, mammalian target of rapamycin (mTOR), and mTOR complex1 (mTORC1). B: Apoptosis induction by decreasing MDM2 expression, a downstream target of Akt, and mTOR, which leads to Bad activation in addition to Bcl-XL and p53 inhibition. C: Induction of autophagy in cancer cells via activating ULK1 after mTOR inhibition, resulting in Beclin-1 phosphorylation. Beclin-1 with AMBRA1 and VPS34 promotes PI3K complex I activity and autophagy in cancer cells. The PI3K/Akt/mTOR pathway overlaps with other intracellular signaling pathways and components such as AMP-activated protein kinase (AMPK), Wnt/β-catenin, mitogen-activated protein kinase (MAPK), and other similar pathways. Cancer therapy can benefit from understanding how these pathways interact and how melatonin affects these pathways.
Collapse
Affiliation(s)
- Vahid Pourbarkhordar
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sohrab Rahmani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Silveira HS, Cesário RC, Vígaro RA, Gaiotte LB, Cucielo MS, Guimarães F, Seiva FRF, Zuccari DAPC, Reiter RJ, Chuffa LGDA. Melatonin changes energy metabolism and reduces oncogenic signaling in ovarian cancer cells. Mol Cell Endocrinol 2024; 592:112296. [PMID: 38844096 DOI: 10.1016/j.mce.2024.112296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Ovarian cancer (OC) adjusts energy metabolism in favor of its progression and dissemination. Because melatonin (Mel) has antitumor actions, we investigated its impact on energy metabolism and kinase signaling in OC cells (SKOV-3 and CAISMOV-24). Cells were divided into control and Mel-treated groups, in the presence or absence of the antagonist luzindole. There was a decrease in the levels of HIF-1α, G6PDH, GAPDH, PDH, and CS after Mel treatment even in the presence of luzindole in both OC cells. Mel treatment also reduced the activity of OC-related enzymes including PFK-1, G6PDH, LDH, CS, and GS whereas PDH activity was increased. Lactate and glutamine levels dropped after Mel treatment. Mel further promoted a reduction in the concentrations of CREB, JNK, NF-kB, p-38, ERK1/2, AKT, P70S6K, and STAT in both cell lines. Mel reverses Warburg-type metabolism and possibly reduces glutaminolysis, thereby attenuating various oncogenic molecules associated with OC progression and invasion.
Collapse
Affiliation(s)
- Henrique Spaulonci Silveira
- Department of Structural and Functional Biology, UNESP - São Paulo State University, Institute of Biosciences, Botucatu, 18618-689, São Paulo, Brazil
| | - Roberta Carvalho Cesário
- Department of Structural and Functional Biology, UNESP - São Paulo State University, Institute of Biosciences, Botucatu, 18618-689, São Paulo, Brazil
| | - Renan Aparecido Vígaro
- Department of Structural and Functional Biology, UNESP - São Paulo State University, Institute of Biosciences, Botucatu, 18618-689, São Paulo, Brazil
| | - Leticia Barbosa Gaiotte
- Department of Structural and Functional Biology, UNESP - São Paulo State University, Institute of Biosciences, Botucatu, 18618-689, São Paulo, Brazil
| | - Maira Smaniotto Cucielo
- Department of Structural and Functional Biology, UNESP - São Paulo State University, Institute of Biosciences, Botucatu, 18618-689, São Paulo, Brazil
| | - Fernando Guimarães
- Hospital da Mulher "Professor Doutor José Aristodemo Pinotti" - CAISM, UNICAMP, Campinas, São Paulo, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Department of Structural and Functional Biology, UNESP - São Paulo State University, Institute of Biosciences, Botucatu, 18618-689, São Paulo, Brazil
| | | | - Russel J Reiter
- Department of Cellular and Structural Biology, UTHealth, San Antonio, TX, 78229, USA
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, UNESP - São Paulo State University, Institute of Biosciences, Botucatu, 18618-689, São Paulo, Brazil.
| |
Collapse
|
3
|
Hosseinzadeh A, Alinaghian N, Sheibani M, Seirafianpour F, Naeini AJ, Mehrzadi S. Melatonin: Current evidence on protective and therapeutic roles in gynecological diseases. Life Sci 2024; 344:122557. [PMID: 38479596 DOI: 10.1016/j.lfs.2024.122557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Melatonin, a potent antioxidant and free radical scavenger, has been demonstrated to be effective in gynecological conditions and female reproductive cancers. This review consolidates the accumulating evidence on melatonin's multifaceted protective effects in different pathological contexts. In gynecological conditions such as endometriosis, polycystic ovary syndrome (PCOS), and uterine leiomyoma, melatonin has shown promising effects in reducing oxidative stress, inflammation, and hormonal imbalances. It inhibits adhesion molecules' production, and potentially mitigates leukocyte adherence and inflammatory responses. Melatonin's regulatory effects on hormone production and insulin sensitivity in PCOS individuals make it a promising candidate for improving oocyte quality and menstrual irregularities. Moreover, melatonin exhibits significant antitumor effects by modulating various signaling pathways, promoting apoptosis, and suppressing metastasis in breast cancers and gynecological cancers, including ovarian, endometrial, and cervical cancers. Furthermore, melatonin's protective effects are suggested to be mediated by interactions with its receptors, estrogen receptors and other nuclear receptors. The regulation of clock-related genes and circadian clock systems may also contribute to its inhibitory effects on cancer cell growth. However, more comprehensive research is warranted to fully elucidate the underlying molecular mechanisms and establish melatonin as a potential therapeutic agent for these conditions.
Collapse
Affiliation(s)
- Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nazila Alinaghian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Ali Jamshidi Naeini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Liao Y, Li R, Pei J, Zhang J, Chen B, Dong H, Feng X, Zhang H, Shang Y, Sui L, Kong Y. Melatonin suppresses tumor proliferation and metastasis by targeting GATA2 in endometrial cancer. J Pineal Res 2024; 76:e12918. [PMID: 37814536 DOI: 10.1111/jpi.12918] [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] [Scholar Register] [Received: 02/27/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023]
Abstract
Endometrial cancer (EC) is a reproductive system disease that occurs in perimenopausal and postmenopausal women. However, its etiology is unclear. Melatonin (MT) has been identified as a therapeutic agent for EC; however, its exact mechanism remains unclear. In the present study, we determined that GATA-binding protein 2 (GATA2) is expressed at low levels in EC and regulated by MT. MT upregulates the expression of GATA2 through MT receptor 1A (MTNR1A), whereas GATA2 can promote the expression of MTNR1A by binding to its promoter region. In addition, in vivo and in vitro experiments showed that MT inhibited the proliferation and metastasis of EC cells by upregulating GATA2 expression. The protein kinase B (AKT) pathway was also affected. In conclusion, these findings suggest that MT and GATA2 play significant roles in EC development.
Collapse
Affiliation(s)
- Yangyou Liao
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ruiling Li
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jingyuan Pei
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Juan Zhang
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Bo Chen
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Haojie Dong
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyu Feng
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Hongshuo Zhang
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yuhong Shang
- Department of Gynecology, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Linlin Sui
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Kong
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
5
|
Cucielo MS, Freire PP, Emílio-Silva MT, Romagnoli GG, Carvalho RF, Kaneno R, Hiruma-Lima CA, Delella FK, Reiter RJ, Chuffa LGDA. Melatonin enhances cell death and suppresses the metastatic capacity of ovarian cancer cells by attenuating the signaling of multiple kinases. Pathol Res Pract 2023; 248:154637. [PMID: 37356221 DOI: 10.1016/j.prp.2023.154637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND Ovarian cancer is a highly aggressive disease that is frequently diagnosed in advanced stages. Melatonin, with its numerous antitumor properties, holds great promise in cancer treatment. Herein, we investigated the effects of melatonin on apoptosis, cell migration, and kinase levels in human ovarian carcinoma SKOV-3 cells and determined whether these effects are mediated by the activation of the MT1 receptor. METHODS SKOV-3 cells were exposed to different concentrations of melatonin based on the presence of MT1 receptor, and we also performed specific silencing of the melatonin receptor gene MTNR1A. RESULTS Our findings revealed that melatonin reduced cell viability as shown by the MTT assay, and flow cytometry analysis showed increased rates of apoptosis and necrosis in all melatonin-treated cells. Melatonin significantly decreased the migratory and invasive capacities of the cells. Propidium iodide labeling indicated that melatonin induced cell cycle arrest by reducing DNA content in the S and G2/M phases in SKOV-3 cells. Additionally, the levels of AKT, ERK1/2, JNK, CREB, p70S6K, STAT3/5, and p38 MAP kinase involved in cell survival, proliferation, motility, and stress responses were depressed by melatonin and further reduced after MT1 knockdown. These molecules were found to be associated with lower overall survival in ovarian cancer patients. CONCLUSIONS Melatonin had obvious oncostatic actions on ovarian cancer cells, and MT1 receptor knockdown intensified its antitumor effect. The inhibition of the MT1 receptor resulted in a substantial reduction in the migratory and invasive capacities of the cells, suggesting its potential as a therapeutic target for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Maira Smaniotto Cucielo
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - Sao Paulo State University, Botucatu 18618-689 São Paulo, Brazil
| | - Paula Paccielli Freire
- Department of Immunology, Institute of Biomedical Sciences, USP - University of São Paulo, São Paulo, Brazil
| | - Maycon Tavares Emílio-Silva
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - Sao Paulo State University, Botucatu 18618-689 São Paulo, Brazil
| | | | - Robson Francisco Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - Sao Paulo State University, Botucatu 18618-689 São Paulo, Brazil
| | - Ramon Kaneno
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - Sao Paulo State University, Botucatu 18618-689 São Paulo, Brazil
| | - Clélia Akiko Hiruma-Lima
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - Sao Paulo State University, Botucatu 18618-689 São Paulo, Brazil
| | - Flávia Karina Delella
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - Sao Paulo State University, Botucatu 18618-689 São Paulo, Brazil
| | - Russel J Reiter
- Departament of Cell Systems and Anatomy, UT Health, San Antonio, TX 782229, USA
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - Sao Paulo State University, Botucatu 18618-689 São Paulo, Brazil.
| |
Collapse
|
6
|
Cosme P, Rodríguez AB, Garrido M, Espino J. Coping with Oxidative Stress in Reproductive Pathophysiology and Assisted Reproduction: Melatonin as an Emerging Therapeutical Tool. Antioxidants (Basel) 2022; 12:antiox12010086. [PMID: 36670948 PMCID: PMC9854935 DOI: 10.3390/antiox12010086] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Infertility is an increasing global public health concern with socio-psychological implications for affected couples. Remarkable advances in reproductive medicine have led to successful treatments such as assisted reproductive techniques (ART). However, the search for new therapeutic tools to improve ART success rates has become a research hotspot. In the last few years, pineal indolamine melatonin has been investigated for its powerful antioxidant properties and its role in reproductive physiology. It is considered a promising therapeutical agent to counteract the detrimental effects associated with oxidative stress in fertility treatments. The aim of the present narrative review was to summarize the current state of the art on the importance of melatonin in reproductive physiology and to provide a critical evaluation of the data available encompassing basic, translational and clinical studies on its potential use in ART to improve fertility success rates.
Collapse
Affiliation(s)
| | | | - María Garrido
- Correspondence: (M.G.); (J.E.); Tel.: +34-924289796 (M.G. & J.E.)
| | - Javier Espino
- Correspondence: (M.G.); (J.E.); Tel.: +34-924289796 (M.G. & J.E.)
| |
Collapse
|
7
|
Mihanfar A, Yousefi B, Azizzadeh B, Majidinia M. Interactions of melatonin with various signaling pathways: implications for cancer therapy. Cancer Cell Int 2022; 22:420. [PMID: 36581900 PMCID: PMC9798601 DOI: 10.1186/s12935-022-02825-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 12/06/2022] [Indexed: 12/30/2022] Open
Abstract
Melatonin is a neuro-hormone with conserved roles in evolution. Initially synthetized as an antioxidant molecule, it has gained prominence as a key molecule in the regulation of the circadian rhythm. Melatonin exerts its effect by binding to cytoplasmic and intra-nuclear receptors, and is able to regulate the expression of key mediators of different signaling pathways. This ability has led scholars to investigate the role of melatonin in reversing the process of carcinogenesis, a process in which many signaling pathways are involved, and regulating these pathways may be of clinical significance. In this review, the role of melatonin in regulating multiple signaling pathways with important roles in cancer progression is discussed, and evidence regarding the beneficence of targeting malignancies with this approach is presented.
Collapse
Affiliation(s)
- Ainaz Mihanfar
- grid.412763.50000 0004 0442 8645Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- grid.412888.f0000 0001 2174 8913Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bita Azizzadeh
- grid.449129.30000 0004 0611 9408Department of Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Maryam Majidinia
- grid.412763.50000 0004 0442 8645Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
8
|
Erdogan CS, Al Hassadi Y, Aru B, Yilmaz B, Gemici B. Combinatorial effects of melatonin and paclitaxel differ depending on the treatment scheme in colorectal cancer in vitro. Life Sci 2022; 308:120927. [PMID: 36063977 DOI: 10.1016/j.lfs.2022.120927] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 10/31/2022]
Abstract
AIMS Colorectal carcinoma (CRC) is the third most prevalent cancer with high mortality. Besides regulating the circadian rhythm, melatonin (MTN) exerts anticancer activities. Paclitaxel (PTX) is successful against different malignancies, however, acquired resistance and variability in patient response restrict its use. mTOR and MAPK pathways are often deregulated in human cancers. We aimed to investigate whether MTN enhances or sensitizes the chemotherapeutic activity of PTX and if so, determine the underlying possible mechanisms in CRC in vitro. MAIN METHODS Antiproliferative and cytotoxic activities of PTX and MTN were assessed alone and in combination, as well as with different treatment regimens (renewal or replacement of the treatment after 24 h), up to 48 h. Apoptosis, viability and autophagy were assessed by flow cytometry. mTOR and MAPK pathway activities were investigated by immunoblotting. KEY FINDINGS Both drugs reduced cell viability in a dose-dependent manner at 24 and 48 h. Only the highest dose of MTN (500 μM) potentiated the cytotoxicity of PTX (50 nM). Replacement of PTX after 24 h with MTN was superior in reducing cell viability than vice versa via apoptosis induction. Renewal of MTN treatment every 24 h reduced autophagy compared to the control group, while other treatments did not alter the autophagic activity. A 24 h MTN treatment followed by 24 h PTX treatment increased S6 phosphorylation in a mTOR-independent manner and increased Erk1/2 phosphorylation. SIGNIFICANCE The present study suggests that sequential treatment with MTN and PTX distinctly affect apoptosis and cytotoxicity via regulating mTOR and MAPK pathways differentially in CRC.
Collapse
Affiliation(s)
- Cihan Suleyman Erdogan
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Yasmine Al Hassadi
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Basak Aru
- Yeditepe University, Faculty of Medicine, Department of Immunology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Bayram Yilmaz
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey
| | - Burcu Gemici
- Yeditepe University, Faculty of Medicine, Department of Physiology, Kayisdagi cad., 34755 Istanbul, Turkey.
| |
Collapse
|
9
|
Lee S, Byun JK, Kim NY, Jin J, Woo H, Choi YK, Park KG. Melatonin inhibits glycolysis in hepatocellular carcinoma cells by downregulating mitochondrial respiration and mTORC1 activity. BMB Rep 2022; 55:459-464. [PMID: 35651333 PMCID: PMC9537022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2022] [Accepted: 03/23/2022] [Indexed: 03/08/2024] Open
Abstract
Various mechanisms have been suggested to explain the chemopreventive and tumor-inhibitory effects of melatonin. Despite the growing evidence supporting melatonin-induced mitochondrial dysfunction, it remains largely unknown how this phenomenon modulates metabolic reprogramming in cancer cells. The aim of our study was to identify the mechanism underlying the anti-proliferative and apoptotic effects of melatonin, which is known to inhibit glycolysis. We analyzed the time-dependent effects of melatonin on mitochondrial respiration and glycolysis in liver cancer cells. The results showed that from a cell bioenergetic point of view, melatonin caused an acute reduction in mitochondrial respiration, however, increased reactive oxygen species production, thereby inhibiting mTORC1 activity from an early stage post-treatment without affecting glycolysis. Nevertheless, administration of melatonin for a longer time reduced expression of c-Myc protein, thereby suppressing glycolysis via downregulation of HK2 and LDHA. The data presented herein suggest that melatonin suppresses mitochondrial respiration and glycolysis simultaneously in HCC cells, leading to anti-cancer effects. Thus, melatonin can be used as an adjuvant agent for therapy of liver cancer. [BMB Reports 2022; 55(9): 459-464].
Collapse
Affiliation(s)
- Seunghyeong Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Jun-Kyu Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| | - Na-Young Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Jonghwa Jin
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Hyein Woo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Yeon-Kyung Choi
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| | - Keun-Gyu Park
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41566, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
10
|
Melatonin Receptors: A Key Mediator in Animal Reproduction. Vet Sci 2022; 9:vetsci9070309. [PMID: 35878326 PMCID: PMC9320721 DOI: 10.3390/vetsci9070309] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 01/26/2023] Open
Abstract
Melatonin, a hormone produced by the mammalian pineal gland, influences various physiological activities, many of which are related to animal reproduction, including neuroendocrine function, rhythm regulation, seasonal behavior, gonadogenesis, gamete development and maturation, sexual maturation, and thermoregulation. Melatonin exerts beneficial actions mainly via binding with G-protein-coupled receptors (GPCR), termed MT1 and MT2. Melatonin receptors are crucial for mediating animal reproduction. This paper reviews the characteristics of melatonin receptors including MT1 and MT2, as well as their roles in mediating signal transduction and biological effects, with a focus on their function in animal reproduction. In addition, we briefly summarize the developments in pharmacological research regarding melatonin receptors as drug targets. It is expected that this review will provide a reference for further exploration and unveiling of melatonin receptor function in reproductive regulation.
Collapse
|
11
|
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] [Scholar 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
|
12
|
Nikolaev G, Robeva R, Konakchieva R. Membrane Melatonin Receptors Activated Cell Signaling in Physiology and Disease. Int J Mol Sci 2021; 23:ijms23010471. [PMID: 35008896 PMCID: PMC8745360 DOI: 10.3390/ijms23010471] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023] Open
Abstract
The pineal hormone melatonin has attracted great scientific interest since its discovery in 1958. Despite the enormous number of basic and clinical studies the exact role of melatonin in respect to human physiology remains elusive. In humans, two high-affinity receptors for melatonin, MT1 and MT2, belonging to the family of G protein-coupled receptors (GPCRs) have been cloned and identified. The two receptor types activate Gi proteins and MT2 couples additionally to Gq proteins to modulate intracellular events. The individual effects of MT1 and MT2 receptor activation in a variety of cells are complemented by their ability to form homo- and heterodimers, the functional relevance of which is yet to be confirmed. Recently, several melatonin receptor genetic polymorphisms were discovered and implicated in pathology-for instance in type 2 diabetes, autoimmune disease, and cancer. The circadian patterns of melatonin secretion, its pleiotropic effects depending on cell type and condition, and the already demonstrated cross-talks of melatonin receptors with other signal transduction pathways further contribute to the perplexity of research on the role of the pineal hormone in humans. In this review we try to summarize the current knowledge on the membrane melatonin receptor activated cell signaling in physiology and pathology and their relevance to certain disease conditions including cancer.
Collapse
Affiliation(s)
- Georgi Nikolaev
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1504 Sofia, Bulgaria;
- Correspondence:
| | - Ralitsa Robeva
- Department of Endocrinology, Faculty of Medicine, Medical University, 1431 Sofia, Bulgaria;
| | - Rossitza Konakchieva
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1504 Sofia, Bulgaria;
| |
Collapse
|
13
|
Lim JO, Lee SJ, Kim WI, Pak SW, Kim JC, Kim JS, Cho YK, Lee IC, Shin IS. Melatonin Alleviates Silica Nanoparticle-Induced Lung Inflammation via Thioredoxin-Interacting Protein Downregulation. Antioxidants (Basel) 2021; 10:antiox10111765. [PMID: 34829636 PMCID: PMC8614841 DOI: 10.3390/antiox10111765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 11/21/2022] Open
Abstract
Silica dioxide nanoparticles (SiONPs) have been increasingly used in various industries; however, this has raised concerns regarding their potential toxicity. SiONPs are also a major component in the Asian sand dust that causes pulmonary diseases among the general public. Melatonin exerts some inhibitory effects against lung inflammation. In this study, we explored the therapeutic properties of melatonin against lung inflammation using an SiONPs-induced lung inflammation murine model and SiONPs-stimulated H292 cells, human airway epithelial cell line, by focusing on the involvement of thioredoxin-interacting protein (TXNIP) in the modulation of the MAPKs/AP-1 axis. We induced an inflammatory response by exposing mouse lungs and the H292 cells to SiONPs and confirmed the anti-inflammatory effect of melatonin. Melatonin inhibited the expression of various inflammatory mediators, including TNF-α, IL-6, and IL-1β, in SiONPs-exposed mice and SiONPs-stimulated H292 cells; this inhibition contributed to a decline in inflammatory cell accumulation in the lung tissues. Furthermore, melatonin treatment decreased the expression of MAPKs and AP-1 by downregulating TXNIP, eventually decreasing the production of SiONPs-induced inflammatory mediators. Overall, these data suggest that melatonin reduces SiONPs-induced lung inflammation by downregulating the TXNIP/MAPKs/AP-1 signalling pathway, thereby supporting the use of melatonin as an effective approach to control SiONPs-induced lung inflammation.
Collapse
Affiliation(s)
- Je-Oh Lim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (J.-O.L.); (S.-J.L.); (W.-I.K.); (S.-W.P.); (J.-C.K.); (J.-S.K.)
| | - Se-Jin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (J.-O.L.); (S.-J.L.); (W.-I.K.); (S.-W.P.); (J.-C.K.); (J.-S.K.)
| | - Woong-Il Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (J.-O.L.); (S.-J.L.); (W.-I.K.); (S.-W.P.); (J.-C.K.); (J.-S.K.)
| | - So-Won Pak
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (J.-O.L.); (S.-J.L.); (W.-I.K.); (S.-W.P.); (J.-C.K.); (J.-S.K.)
| | - Jong-Choon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (J.-O.L.); (S.-J.L.); (W.-I.K.); (S.-W.P.); (J.-C.K.); (J.-S.K.)
| | - Joong-Sun Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (J.-O.L.); (S.-J.L.); (W.-I.K.); (S.-W.P.); (J.-C.K.); (J.-S.K.)
| | - Young-Kwon Cho
- College of Health Sciences, Cheongju University, Chungbuk 28503, Korea;
| | - In-Chul Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeonbuk 56212, Korea
- Correspondence: (I.-C.L.); (I.-S.S.); Tel.: +82-63-570-5239 (I.-C.L.); +82-62-530-2835 (I.-S.S.)
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (J.-O.L.); (S.-J.L.); (W.-I.K.); (S.-W.P.); (J.-C.K.); (J.-S.K.)
- Correspondence: (I.-C.L.); (I.-S.S.); Tel.: +82-63-570-5239 (I.-C.L.); +82-62-530-2835 (I.-S.S.)
| |
Collapse
|
14
|
Tan Y, Li L, Liu H, Yu J, Wang Q, Lin Q. Chinese Medicine Leptochloa chinensis Inhibits the Malignant Behaviors of Renal Cell Carcinoma 786-O Cells by Regulating the mTOR Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:5122380. [PMID: 34675984 PMCID: PMC8526199 DOI: 10.1155/2021/5122380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a common malignant tumor of the urinary system that seriously threatens human life and health. This study aims to explore the role of the traditional Chinese medicine Leptochloa chinensis in the pathogenesis of RCC. Meanwhile, this study also revealed the molecular biological mechanism of its antitumor activity. METHODS Human RCC 786-O cells were cultured in the RPMI-1640 medium, which contains different concentrations of Leptochloa chinensis (1,000, 3,000, and 9,000 μg/ml). MTT and flow cytometry assays were used to detect the viability of 786-O cells. Transwell and wound healing assays were used to detect cell metastasis. The protein expression was observed by western blot analysis. RESULTS Leptochloa can inhibit cell proliferation and induce apoptosis in RCC 786-O cells. In addition, Leptochloa can weaken the migration and invasion of 786-O cells. More importantly, Leptochloa can block the mTOR pathway by inhibiting the protein expression of p-mTOR. Moreover, the high concentration of Leptochloa chinensis has a better inhibitory effect on 786-O cells. CONCLUSION The traditional Chinese medicine Leptochloa chinensis inhibits the viability and metastasis of 786-O cells by blocking the mTOR pathway.
Collapse
Affiliation(s)
- Yongshun Tan
- Department of Nephrology, Jinan City People's Hospital, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan 271199, Shandong Province, China
| | - Lingyun Li
- Department of Internal Medicine, Laishan Branch Hospital of Yantai Yuhuangding Hospital, Yantai, 264003, Shandong Province, China
| | - Hongyue Liu
- Disinfection Supply Center, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, 266000, Shandong Province, China
| | - Jiadong Yu
- Personnel Section, Zhangqiu District People's Hospital, Jinan, 250200, Shandong Province, China
| | - Qijun Wang
- Department of Otorhinolaryngology, Zhangqiu District People's Hospital, Jinan 250200, Shandong Province, China
| | - Qiuju Lin
- Department of Oncology (II), Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, 266000, Shandong Province, China
| |
Collapse
|
15
|
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] [Scholar 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
|
16
|
Yu J, Hu X, Chen X, Zhou Q, Jiang Q, Shi Z, Zhu H. CNOT7 modulates biological functions of ovarian cancer cells via AKT signaling pathway. Life Sci 2021; 268:118996. [PMID: 33412213 DOI: 10.1016/j.lfs.2020.118996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022]
Abstract
AIMS CNOT7 plays an important role in many biological processes, providing attractive opportunities for the treatment of malignant tumors. However, the functions and mechanism of CNOT7 in ovarian cancer (OC) have not been elucidated. The purpose of this study was to assess the role of CNOT7 in OC. MATERIALS AND METHODS SKOV3 and A2780 cells were chosen as the cell lines for the experiments of this manuscript via the analysis of the expression of CNOT7 protein and the mRNA level in ovarian surface epithelium (OSE) cells, SKOV3, HO8910 and A2780 cells. The expression of CNOT7 was detected by western blot assays and RT-PCR in A2780 and SKOV3 cells. The MTT assays, colony formation assays and EdU assays were used to measure cell proliferation when CNOT7 was knocked down or overexpressed in A2780 and SKOV3 cells. Furthermore, cell migration and invasion ability were achieved from transwell assays. Cell cycle and apoptosis rate after small interference RNA-CNOT7 (siRNA-CNOT7) were detected by flow cytometry assays. Finally, the cell proliferation, migration and invasion ability were detected when A2780 and SKOV3 cells with CNOT7 overexpression were treated with LY294002. KEY FINDINGS The expression of CNOT7 protein in OC cells, including SKOV3, HO8910 and A2780 cells were significantly higher than that in OSE cells (P < 0.05). The mRNA level of CNOT7 in HO8910 and A2780 cells were significantly higher than that in OSE cells (P < 0.01). However, the mRNA level of CNOT7 in SKOV3 cells was no significant difference compared with OSE cells (P > 0.05). The results suggested that knockdown of CNOT7 could inhibit the cell proliferation, migration and invasion ability in A2780 and SKOV3 cells, and increase cell apoptosis and autophagy. The expression of apoptosis-related molecules (PARP, Caspase3 and Caspase9) and autophagy-related protein (LC3B) were up-regulated after CNOT7 knockdown, while the expression of cycle-related protein (CDK6) and the anti-apoptotic gene (Bcl2) were downregulated. Meanwhile, the opposite results were observed when CNOT7 was overexpressed in A2780 and SKOV3 cells. It is worth noting that the effect of CNOT7 overexpression in A2780 and SKOV3 cells could be partially or completely eliminated by treatment with AKT inhibitor LY294002. SIGNIFICANCE CNOT7 has a carcinogenic effect in OC, and the carcinogenic effect may be achieved via the AKT signaling pathway.
Collapse
Affiliation(s)
- Jiangtao Yu
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Xiaoli Hu
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Xiuxiu Chen
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Qiangyong Zhou
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Qi Jiang
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China
| | - Zhengzheng Shi
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China.
| | - Haiyan Zhu
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, People's Republic of China; Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200126, People's Republic of China.
| |
Collapse
|
17
|
Melatonin suppresses chronic restraint stress-mediated metastasis of epithelial ovarian cancer via NE/AKT/β-catenin/SLUG axis. Cell Death Dis 2020; 11:644. [PMID: 32811805 PMCID: PMC7435194 DOI: 10.1038/s41419-020-02906-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 12/27/2022]
Abstract
Chronic stress has been shown to facilitate progression of epithelial ovarian cancer (EOC), however, the neuro-endocranial mechanism participating in this process still remains unclear. Here, we reported that chronic restraint stress (CRS) promoted the abdominal implantation metastasis of EOC cells and the expression of epithelial–mesenchymal transition-related markers in tumor-bearing mouse model, including TWIST, SLUG, SNAIL, and β-catenin. We observed that β-catenin co-expressed with SLUG and norepinephrine (NE) in tumor tissues obtained from nude mice. Further ex vivo experiments revealed that NE promoted migration and invasion of ovarian cancer cells and SLUG expression through upregulating expression and improving transcriptional function of β-catenin in vitro. A human phosphor-kinase array suggested that NE activated various kinases in ovarian cancer cells, and we further confirmed that AKT inhibitor reduced NE-mediated pro-metastatic impacts and activation of the β-catenin/SLUG axis. Furthermore, the expression levels of NE and β-catenin were examined in ovarian tumor tissues by using tumor tissue arrays. Results showed that the expression levels of both NE and β-catenin were associated with poor clinical stage of serous EOC. Moreover, we found that melatonin (MLT) effectively reduced the abdominal tumor burden of ovarian cancer induced by CRS, which was partially related to the inhibition of the NE/AKT/β-catenin/SLUG axis. Collectively, these findings suggest a novel mechanism for CRS-mediated ovarian cancer metastasis and MLT has a potential therapeutic efficacy against ovarian cancer.
Collapse
|
18
|
Liu K, Xue B, Bai G, Zhang W. F-box protein FBXO31 modulates apoptosis and epithelial-mesenchymal transition of cervical cancer via inactivation of the PI3K/AKT-mediated MDM2/p53 axis. Life Sci 2020; 259:118277. [PMID: 32800832 DOI: 10.1016/j.lfs.2020.118277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022]
Abstract
AIMS Cervical cancer (CC) is one of the most common malignant tumours in the world and a serious threat to women's health. The dysregulation of protein degradation mediated by F-box proteins is involved in tumorigenesis, and F-box protein FBXO31 has been reported to play an important role in various human cancers. However, the role of FBXO31 in CC remains unclear. This study aimed to investigate the function and underlying regulatory mechanism of FBXO31 in CC. MAIN METHODS In this study, quantitative real-time polymerase chain reaction (qRT-PCR) and western blot were used to measure target gene expression; the Cell Counting Kit-8, cell death ELISA, Transwell invasion assay, wound-healing assay and western blot were applied to assess cell viability, apoptosis, invasion, migration and epithelial-mesenchymal transition (EMT), respectively. KEY FINDINGS FBXO31 was expressed at a low level in 37 pairs of CC tissues and three types of CC cell lines. Overexpression of FBXO31 inhibited cell viability, invasion, migration, EMT and induced apoptosis in SiHa cells. FBXO31 promoted p53 activity through suppression of murine double minute 2 (MDM2) expression. Overexpression of MDM2 ameliorated the inhibitory effect of FBXO31 on SiHa cells, while the MDM2/p53 axis-specific inhibitor Nutlin-3a facilitated this inhibitory effect. Further, we confirmed that FBXO31 inactivated MDM2/p53 axis dependence on the phospholipid inositol 3-kinase (PI3K)/protein kinase B (AKT) signalling pathway. SIGNIFICANCE Collectively, our results reveal that FBXO31 down-regulates CC progression by blocking the PI3K/AKT-mediated MDM2/p53 axis, suggesting that FBXO31 may serve as a promising therapeutic target for CC treatment.
Collapse
Affiliation(s)
- Keying Liu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; North Hospital Affiliated to Xi'an Medical College of Xi'an, Xi'an, Shaanxi 710043, China
| | - Biyun Xue
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Guiqin Bai
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Wentao Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| |
Collapse
|
19
|
Samanta S. Melatonin: an endogenous miraculous indolamine, fights against cancer progression. J Cancer Res Clin Oncol 2020; 146:1893-1922. [PMID: 32583237 DOI: 10.1007/s00432-020-03292-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Melatonin is an amphipathic indolamine molecule ubiquitously present in all organisms ranging from cyanobacteria to humans. The pineal gland is the site of melatonin synthesis and secretion under the influence of the retinohypothalamic tract. Some extrapineal tissues (skin, lens, gastrointestinal tract, testis, ovary, lymphocytes, and astrocytes) also enable to produce melatonin. Physiologically, melatonin regulates various functions like circadian rhythm, sleep-wake cycle, gonadal activity, redox homeostasis, neuroprotection, immune-modulation, and anticancer effects in the body. Inappropriate melatonin secretion advances the aging process, tumorigenesis, visceral adiposity, etc. METHODS: For the preparation of this review, I had reviewed the literature on the multidimensional activities of melatonin from the NCBI website database PubMed, Springer Nature, Science Direct (Elsevier), Wiley Online ResearchGate, and Google Scholar databases to search relevant articles. Specifically, I focused on the roles and mechanisms of action of melatonin in cancer prevention. RESULTS The actions of melatonin are primarily mediated by G-protein coupled MT1 and MT2 receptors; however, several intracellular protein and nuclear receptors can modulate the activity. Normal levels of the melatonin protect the cells from adverse effects including carcinogenesis. Therapeutically, melatonin has chronomedicinal value; it also shows a remarkable anticancer property. The oncostatic action of melatonin is multidimensional, associated with the advancement of apoptosis, the arrest of the cell cycle, inhibition of metastasis, and antioxidant activity. CONCLUSION The present review has emphasized the mechanism of the anti-neoplastic activity of melatonin that increases the possibilities of the new approaches in cancer therapy.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department Physiology, Midnapore College, Paschim Medinipur, Midnapore, West Bengal, 721101, India.
| |
Collapse
|
20
|
Neuroprotection of melatonin on spinal cord injury by activating autophagy and inhibiting apoptosis via SIRT1/AMPK signaling pathway. Biotechnol Lett 2020; 42:2059-2069. [PMID: 32514788 DOI: 10.1007/s10529-020-02939-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/05/2020] [Indexed: 12/23/2022]
Abstract
The effect of melatonin (MT) on spinal cord injury (SCI) has attracted increasing research attention. However, the specific role and molecular mechanism of MT on SCI have not been elucidated. An experiment was performed to investigate the effect and molecular mechanism of MT on the neuronal autophagy after SCI and its effect on the recovery of nerve function. The rats were randomly divided into four treatment groups: the SCI+MT+EX527 (SIRT1 inhibitor), SCI+MT, SCI, and sham operation groups. On the 14th day after SCI, MT significantly promoted the recovery of motor function in the hind limbs according to the results of Basso, Beattie, and Bresnahan scores. At the same time, MT treatment resulted in reduced activation of cleaved-caspase-3, cleaved-caspase-9, and terminal deoxynucleotidyl transferase dUTP nick end labeling-positive neurons and increased the survival of motoneurons in the anterior horn of the spinal cord on the 14th day after SCI, which exerted its neuroprotection. Furthermore, western blot and immunofluorescence double staining were performed to verify the molecular mechanism of effect of MT on SCI, and results showed the significantly upregulated expressions of Beclin-1, light chain-3B, SIRT1, p-AMPK proteins in the spinal cord tissue of MT-treated rats on the 14th day after SCI, however, the effect of MT on autophagy was reversed by EX527 (SIRT1 inhibitor), which implied that MT activated autophagy via SIRT1/AMPK signaling pathway after SCI. Similarly, the neuroprotective effects of MT on SCI were also inhibited after the SIRT1/AMPK signaling pathway was suppressed by EX527. Taken together, these results suggest that MT inhibits the apoptosis and activates autophagy of nerve cells after SCI and ultimately exerts the neuroprotective effect by SIRT1/AMPK signaling pathway.
Collapse
|
21
|
Kasi R, Yeo PL, Yen NK, Koh RY, Ponnudurai G, Tiong YL, Chye SM. Melatonin Induces Apoptosis and Inhibits the Proliferation of Cancer Cells via Reactive Oxygen Species-mediated MAPK and mTOR Pathways. ACTA ACUST UNITED AC 2020. [DOI: 10.2174/2212697x06666191116151114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Background:
Recent human and animal studies have demonstrated the oncostatic properties
of N-acetyl-5-methoxytryptamine (melatonin) in different types of cancer. However, in few cancer
cell lines including colorectal cancer cell line (HT-29), acute T cell leukemia cell line (JURKAT)
and cervical cancer cell line (HeLa), precise oncostatic mechanism induced by melatonin is yet to be
described.
Objectives:
The aim of this study is to investigate the effects of melatonin in HT-29, JURKAT and
HeLa cells and to determine the underlying molecular mechanism.
Methods:
Cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
(MTT) assay while cell cycle, apoptosis and membrane potential were analysed by flow cytometry.
Reactive oxygen species (ROS) was detected by 2',7'.dichlorofluorescein diacetate(DCFH-DA)
staining. Protein expressions were determined by Western blot.
Results:
Our results showed that melatonin suppressed cell proliferation, increased the number of sub
G1 hypodiploid cells and cell cycle arrest in HT-29, JURKAT and HeLa cells. Besides, melatonin also
induced early and late apoptosis, although there were marked variations in responses between different
cell lines (sensitivity; HeLa > HT-29 >JURKAT). Apart from that, staining with DCHF-DA
demonstrated ROS production that was induced in a dose-dependent manner in HeLa, HT-29 and
JURKAT cells. Moreover, the apoptotic process and oncostatic effect of melatonin were seen to be
associated with extracellular-signal-regulated kinase (ERK) and stress-activated protein kinase/c-Jun
NH (2)-terminal kinase (SAPK-JNK) signalling cascades in HeLa cells. In HT-29 and JURKAT cells,
melatonin induced apoptosis via activation of p38 mitogen-activated protein kinases (p38), ERK and
SAPK-JNK signalling pathways. In all three cell lines, the apoptotic event was triggered by the
mammalian target of rapamycin (mTOR)-mediated activation of the downstream target rapamycininsensitive
companion of mTOR (RICTOR) and/or regulatory-associated protein of mTOR (RAPTOR)
proteins.
Conclusions:
Our findings confirm that melatonin induces apoptosis through reactive oxygen speciesmediated
dysregulated mitogen-activated protein kinase (MAPK) and mTOR signalling pathways in
these cancer cell lines.
Collapse
Affiliation(s)
- Reena Kasi
- School of Postgraduate, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Pei Ling Yeo
- School of Postgraduate, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Ng. Khuen Yen
- School of Pharmacy, Monash University Malaysia, Selangor 47500, Malaysia
| | - Rhun Yian Koh
- School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | | | - Yee Lian Tiong
- School of Postgraduate, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Soi Moi Chye
- School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
22
|
Zhu H, Chen Y, Bai LC, Cao XR, Xu R. Different Effects of Melatonin on X-Rays-Irradiated Cancer Cells in a Dose-Dependent Manner. Dose Response 2019; 17:1559325819877271. [PMID: 31579126 PMCID: PMC6759722 DOI: 10.1177/1559325819877271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/18/2018] [Accepted: 08/20/2019] [Indexed: 11/17/2022] Open
Abstract
The purpose of this study is to investigate the effects of melatonin on the radiosensitivity of HeLa cells. Concentration from 10 to 1000 µM of melatonin was used on HeLa cells before X-rays irradiation (IR). The cellular inactivation effect was analyzed by clonogenic assay, and cell growth was measured by MTT assay at various concentrations. Ten micrometer melatonin promoted the cell-killing effects of IR, while 1000-µM melatonin prevented IR-induced cellular inactivation. Further analysis revealed that 1000-µM melatonin protected the cells from IR-induced reactive oxygen species damage, as the oxidative stress measured by fluorescent microscopy and fluorescence-activated cell sorting using 2,7-dichlorofluorescein diacetate staining. This is further confirmed by melatonin receptor agonist, which has no antioxidant capacity. A 10-µM melatonin, on the contrary, enhanced the cell-killing effects of IR by activating c-Jun NH2-terminal kinase (JNK) signaling. c-Jun NH2-terminal kinase signaling activation was indicated by Western blot of phosphorylated JNK. We used JNK inhibitor to further confirm the involvement of JNK signaling in the cell-killing enhancement of 10-µM melatonin administration. Our results suggest the importance of dose-dependent effects in melatonin application for radiotherapy.
Collapse
Affiliation(s)
- Hao Zhu
- Radiology Department, Lanzhou University Second Hospital, Lanzhou, China
| | - Yong Chen
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Liang-Cai Bai
- Radiology Department, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiang-Rong Cao
- Radiology Department, Lanzhou University Second Hospital, Lanzhou, China
| | - Rui Xu
- Radiology Department, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
23
|
Genario R, Morello E, Bueno AA, Santos HO. The usefulness of melatonin in the field of obstetrics and gynecology. Pharmacol Res 2019; 147:104337. [PMID: 31276773 DOI: 10.1016/j.phrs.2019.104337] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/28/2019] [Accepted: 06/28/2019] [Indexed: 01/24/2023]
Abstract
Disorders of the female reproductive system, including those associated with hormone regulation, fertility rate and fetal health, are issues of great concern worldwide. More recently, melatonin supplementation has been suggested as a therapeutic approach in gynecological practice. In both animal models and in women, melatonin supplementation suggests a therapeutic and preventative potential, effects attributed mainly to its antioxidant properties and action as hormone modulator. The aim of this literature review is to further investigate the evidence available on the effects of melatonin supplementation in animal and human studies, focusing on its potential application to gynecology. Melatonin-containing supplements are easily found in online and high street retailers, and despite its supplementation deemed to be relatively safe, no consensus has been reached on effective dosage and supplementation period. Short term supplementation studies, of up to six months, suggest that a daily posology of 2-18 mg of melatonin may have the potential to improve fertility rate, oocyte quality, maturation and number of embryos. However, the evidence available so far on the effects of melatonin supplementation covering gestational age and gestational outcomes is very scarce. Clinical trials and longer-term supplementation studies are required to assess any clinical outcome associated with melatonin supplementation in the field of gynecology.
Collapse
Affiliation(s)
- Rafael Genario
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil.
| | | | - Allain Amador Bueno
- Department of Biological Sciences, University of Worcester, Henwick Grove, Worcester, WR2 6AJ, United Kingdom.
| | - Heitor Oliveira Santos
- School of Medicine, Federal University of Uberlandia (UFU), Av. Para, nº1720 Bloco 2U Campus Umuarama, Uberlandia, Minas Gerais, 38400-902, Brazil.
| |
Collapse
|
24
|
Jafari‐Vayghan H, Saleh‐Ghadimi S, Maleki V, Moludi J, Alizadeh M. The effects of melatonin on neurohormonal regulation in cardiac cachexia: A mechanistic review. J Cell Biochem 2019; 120:16340-16351. [DOI: 10.1002/jcb.29151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Hamed Jafari‐Vayghan
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science Tabriz University of Medical Sciences Tabriz Iran
- Nutrition Research Center, Faculty of Nutrition and Food Sciences Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Sevda Saleh‐Ghadimi
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Vahid Maleki
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Jalal Moludi
- Department of Nutrition, Faculty of Nutrition Sciences and Food Technology Kermanshah University of Medical Sciences Kermanshah Iran
| | - Mohammad Alizadeh
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science Tabriz University of Medical Sciences Tabriz Iran
- Nutrition Research Center, Faculty of Nutrition and Food Sciences Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
25
|
Zare H, Shafabakhsh R, Reiter RJ, Asemi Z. Melatonin is a potential inhibitor of ovarian cancer: molecular aspects. J Ovarian Res 2019; 12:26. [PMID: 30914056 PMCID: PMC6434863 DOI: 10.1186/s13048-019-0502-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer is one of the most common causes of morbidity related to gynecologic malignancies. Possible risk factors are including hereditary ovarian cancer, obesity, diabetes mellitus, alcohol consumption, aging, and smoking. Various molecular signaling pathways including inflammation, oxidative stress, apoptosis and angiogenesis are involved in this progression of ovarian cancer. Standard treatments for recently diagnosed patients are Surgery and chemotherapy such as co-treatment with other drugs such that the exploitation of neoadjuvant chemotherapy is expanding. Melatonin (N-acetyl-5-methoxy-tryptamine), an endogenous agent secreted from the pineal gland, has anti-carcinogenic features, such as regulation of estradiol production, cell cycle modulation, stimulation of apoptosis as well as anti-angiogenetic properties, anti-inflammatory activities, significant antioxidant effects and modulation of various immune system cells and cytokines. Multiple studies have shown the significant beneficial roles of melatonin in various types of cancers including ovarian cancer. This paper aims to shed light on the roles of melatonin in ovarian cancer treatment from the standpoint of the molecular aspects.
Collapse
Affiliation(s)
- Hadis Zare
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R, Iran
| | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R, Iran
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science, Center, San Antonio, TX, USA
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R, Iran.
| |
Collapse
|
26
|
Silva TM, Moretto FCF, Sibio MTD, Gonçalves BM, Oliveira M, Olimpio RMC, Oliveira DAM, Costa SMB, Deprá IC, Namba V, Nunes MT, Nogueira CR. Triiodothyronine (T3) upregulates the expression of proto-oncogene TGFA independent of MAPK/ERK pathway activation in the human breast adenocarcinoma cell line, MCF7. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:142-147. [PMID: 30916164 PMCID: PMC10522138 DOI: 10.20945/2359-3997000000114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 12/12/2018] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To verify the physiological action of triiodothyronine T3 on the expression of transforming growth factor α (TGFA) mRNA in MCF7 cells by inhibition of RNA Polymerase II and the MAPK/ERK pathway. MATERIALS AND METHODS The cell line was treated with T3 at a physiological dose (10-9M) for 10 minutes, 1 and 4 hour (h) in the presence or absence of the inhibitors, α-amanitin (RNA polymerase II inhibitor) and PD98059 (MAPK/ERK pathway inhibitor). TGFA mRNA expression was analyzed by RT-PCR. For data analysis, we used ANOVA, complemented with the Tukey test and Student t-test, with a minimum significance of 5%. RESULTS T3 increases the expression of TGFA mRNA in MCF7 cells in 4 h of treatment. Inhibition of RNA polymerase II modulates the effect of T3 treatment on the expression of TGFA in MCF7 cells. Activation of the MAPK/ERK pathway is not required for T3 to affect the expression of TGFA mRNA. CONCLUSION Treatment with a physiological concentration of T3 after RNA polymerase II inhibition altered the expression of TGFA. Inhibition of the MAPK/ERK pathway after T3 treatment does not interfere with the TGFA gene expression in a breast adenocarcinoma cell line.
Collapse
Affiliation(s)
- Tabata M. Silva
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Fernanda C. F. Moretto
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Maria T. De Sibio
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Bianca M. Gonçalves
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Miriane Oliveira
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Regiane M. C. Olimpio
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Diego A. M. Oliveira
- Universidade Estadual PaulistaUniversidade Estadual PaulistaBotucatuSPBrasilUniversidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Sarah M. B. Costa
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Igor C. Deprá
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Vickeline Namba
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| | - Maria T. Nunes
- Universidade de São PauloUniversidade de São PauloInstituto de Ciências BiomédicasDepartamento de Fisiologia e BiofísicaSão PauloSPBrasilDepartamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brasil
| | - Célia R. Nogueira
- Universidade Estadual PaulistaUniversidade Estadual PaulistaFaculdade de Medicina de BotucatuDepartamento de Medicina InternaBotucatuSPBrasilDepartamento de Medicina Interna, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista (UNESP), Botucatu, SP, Brasil
| |
Collapse
|
27
|
Tan GX, Wang XN, Tang YY, Cen WJ, Li ZH, Wang GC, Jiang JW, Wang XC. PP-22 promotes autophagy and apoptosis in the nasopharyngeal carcinoma cell line CNE-2 by inducing endoplasmic reticulum stress, downregulating STAT3 signaling, and modulating the MAPK pathway. J Cell Physiol 2019; 234:2618-2630. [PMID: 30191969 DOI: 10.1002/jcp.27076] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/28/2018] [Indexed: 12/24/2022]
Abstract
Paris polyphylla var. yunnanensis, named Chong Lou, is considered an antitumor substance. In this study, we investigated the effect of PP-22, a monomer purified from P. polyphylla var. yunnanensis, on the nasopharyngeal carcinoma cell line CNE-2 in vitro. The results showed that PP-22 could inhibit the proliferation of CNE-2 cells via the induction of apoptosis, with evidence of the characteristic morphological changes in the apoptosis in the nucleus and an increase in Annexin V-positive cells. In addition, we found that PP-22 could activate the p38 mitogen-activated protein kinase (MAPK) pathway and that this activation was reversed by SB203580, a specific inhibitor of the p38 MAPK pathway. In contrast, PP-22 promoted apoptosis via an intrinsic pathway, including the endoplasmic reticulum stress pathway, in a caspase-dependent manner. A further study showed that PP-22 also induced apoptosis by downregulating the signal transducers and activators of transcription 3 (STAT3) pathway, and the inhibitory effect was also confirmed by STAT3 small interfering RNA. In addition, PP-22 could promote autophagy by inhibiting the extracellular regulated protein kinases (ERK) pathway. And autophagy plays a protective role against apoptosis. Together, these data show that PP-22 promotes autophagy and apoptosis in the nasopharyngeal carcinoma CNE-2 cell line.
Collapse
Affiliation(s)
- Gui-Xiang Tan
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Gastroenterology, People's Hospital of Qingyuan, Guangdong, China
| | - Xin-Ning Wang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yun-Yun Tang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Wan-Jing Cen
- Department of Stomatology, Guangzhou Development District Hospital, Guangzhou, China
| | - Zhen-Hua Li
- Translation Research Institute, Jinan University, Guangzhou, China
| | - Guo-Cai Wang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jian-Wei Jiang
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, China
| | - Xi-Cheng Wang
- Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
28
|
Menéndez-Menéndez J, Martínez-Campa C. Melatonin: An Anti-Tumor Agent in Hormone-Dependent Cancers. Int J Endocrinol 2018; 2018:3271948. [PMID: 30386380 PMCID: PMC6189685 DOI: 10.1155/2018/3271948] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/30/2018] [Accepted: 08/12/2018] [Indexed: 02/07/2023] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is a hormone synthesized and secreted by the pineal gland mainly during the night, since light exposure suppresses its production. Initially, an implication of this indoleamine in malignant disease was described in endocrine-responsive breast cancer. Data from several clinical trials and multiple experimental studies performed both in vivo and in vitro have documented that the pineal hormone inhibits endocrine-dependent mammary tumors by interfering with the estrogen signaling-mediated transcription, therefore behaving as a selective estrogen receptor modulator (SERM). Additionally, melatonin regulates the production of estradiol through the control of the enzymes involved in its synthesis, acting as a selective estrogen enzyme modulator (SEEM). Many more mechanisms have been proposed during the past few years, including signaling triggered after activation of the membrane melatonin receptors MT-1 and MT-2, or else intracellular actions targeting molecules such as calmodulin, or binding intranuclear receptors. Similar results have been obtained in prostate (regulation of enzymes involved in androgen synthesis and modulation of androgen receptor levels and activity) and ovary cancer. Thus, tumor metabolism, gene expression, or epigenetic modifications are modulated, cell growth is impaired and angiogenesis and metastasis are inhibited. In the last decade, many more reports have demonstrated that melatonin is a promising adjuvant molecule with many potential beneficial consequences when included in chemotherapy or radiotherapy protocols designed to treat endocrine-responsive tumors. Therefore, in this state-of-the-art review, we aim to compile the knowledge about the oncostatic actions of the indoleamine in hormone-dependent tumors, and the latest findings concerning melatonin actions when administered in combination with radio- or chemotherapy in breast, prostate, and ovary cancers. As melatonin has no toxicity, it may be well deserve to be considered as an endogenously generated agent helpful in cancer prevention and treatment.
Collapse
Affiliation(s)
- Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| |
Collapse
|
29
|
Lu KH, Su SC, Lin CW, Hsieh YH, Lin YC, Chien MH, Reiter RJ, Yang SF. Melatonin attenuates osteosarcoma cell invasion by suppression of C-C motif chemokine ligand 24 through inhibition of the c-Jun N-terminal kinase pathway. J Pineal Res 2018; 65:e12507. [PMID: 29766567 DOI: 10.1111/jpi.12507] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022]
Abstract
Osteosarcoma, with its high metastatic potential, is the most prevalent malignant bone tumor in children and adolescents. Melatonin possesses multiple tumor-suppressing properties for a myriad of tumors, but little is known about the effects of melatonin on osteosarcoma metastasis. In this study, we demonstrated that melatonin elicited very low cytotoxicity and significantly inhibited cellular motility, migration, and invasion in human osteosarcoma U2OS and HOS cells. Moreover, using RNA sequencing technology, we revealed that melatonin repressed C-C motif chemokine ligand 24 (CCL24) gene expression in U2OS cells. Manipulation of CCL24 levels influenced the motility of osteosarcoma cells as cell migration and invasion were enhanced by the addition of recombinant human CCL24 and attenuated by the silencing of CCL24. Moreover, melatonin increased and decreased the activation of extracellular signal-regulated kinase (ERK) 1/2 and c-Jun N-terminal kinase (JNK) 1/2, respectively, in a dose-dependent manner in U2OS and HOS cells while exerting no evident influence on the level and activation of p38, Akt, FAK, steroid receptor coactivator, or Raf. In further functional experiments, the use of JNK inhibitors (SP600125 and DN-JNK) confirmed that the pharmaceutic inhibition of JNK augmented the melatonin-mediated CCL24 suppression and migration of U2OS cells. Overall, our results revealed that melatonin attenuated chemokine CCL24 levels through inhibition of the JNK pathway to hinder human osteosarcoma cell invasion, thereby highlighting the therapeutic potential of melatonin for osteosarcoma metastasis.
Collapse
Affiliation(s)
- Ko-Hsiu Lu
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Linkou and Keelung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Ya-Chiu Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Russel J Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
30
|
Zhang S, Liu S, Zhang J, Reiter RJ, Wang Y, Qiu D, Luo X, Khalid AR, Wang H, Feng L, Lin Z, Ren M. Synergistic anti-oomycete effect of melatonin with a biofungicide against oomycetic black shank disease. J Pineal Res 2018; 65:e12492. [PMID: 29575191 DOI: 10.1111/jpi.12492] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 03/09/2018] [Indexed: 01/19/2023]
Abstract
Human health, food safety, and agriculture have been threatened by oomycetic diseases caused by notorious pathogenic oomycetes. Chemical oomyceticides are the main approaches in control of pathogenic oomycetes. However, the overused chemical oomyceticides have resulted in serious environmental pollution and drug resistance. The eco-friendly bio-oomyceticides are required for sustainable development through screening synergistic drug combinations. In this study, Phytophthora nicotianae (P. nicotianae), as one of the most destructive oomycetic diseases in agriculture, was used as a model system to screen the novel bio-oomyceticides based on drug combination. The results showed that treatment of melatonin or ethylicin (IUPAC Name: 1-ethylsulfonylsulfanylethane) alone displayed similar phenotypes such as the inhibition of the hyphal growth, reduction of the cell viability, and suppression of the virulence of P. nicotianae. Importantly, melatonin and ethylicin shared the same targets of interfering with the amino acid metabolism, overexpressing apoptosis-inducing factor, and dysregulating the virulence-related genes. Furthermore, strong synergism against P. nicotianae was induced by combining melatonin with ethylicin. Under treatment of the combination of melatonin and ethylicin, the expression of genes associated with amino acid, the apoptosis-inducing factor, and the virulence-related genes was much more significantly dysregulated than that of single drug treatment. Thus, the tobacco black shank caused by P. nicotianae can be successfully controlled using the combination of melatonin and ethylicin. These observations suggest that the synergistic effect based on the combination of melatonin and ethylicin is an eco-friendly alternative for the control of the destructive oomycetic diseases.
Collapse
Affiliation(s)
- Shumin Zhang
- School of Life Sciences, Chongqing University, Chongqing, China
- School of Basic Medical Sciences, North Sichuan Medical College, Nanchong, China
| | - Sen Liu
- College of Agronomy and Biotechnology, Southwest University, Chongqing, China
| | - Jiankui Zhang
- College of Agronomy and Biotechnology, Southwest University, Chongqing, China
| | - Russel J Reiter
- Department of Cellular and Structure Biology, UT Health, San Antonio, TX, USA
| | - Ying Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Dan Qiu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xiumei Luo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - A Rehman Khalid
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Hanyan Wang
- School of Basic Medical Sciences, North Sichuan Medical College, Nanchong, China
| | - Li Feng
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Maozhi Ren
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
31
|
Cecon E, Oishi A, Jockers R. Melatonin receptors: molecular pharmacology and signalling in the context of system bias. Br J Pharmacol 2018; 175:3263-3280. [PMID: 28707298 PMCID: PMC6057902 DOI: 10.1111/bph.13950] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 12/15/2022] Open
Abstract
Melatonin, N-acetyl-5-methoxytryptamine, an evolutionally old molecule, is produced by the pineal gland in vertebrates, and it binds with high affinity to melatonin receptors, which are members of the GPCR family. Among the multiple effects attributed to melatonin, we will focus here on those that are dependent on the activation of the two mammalian MT1 and MT2 melatonin receptors. We briefly summarize the latest developments on synthetic melatonin receptor ligands, including multi-target-directed ligands, and the characterization of signalling-biased ligands. We discuss signalling pathways activated by melatonin receptors that appear to be highly cell- and tissue-dependent, emphasizing the impact of system bias on the functional outcome. Different proteins have been demonstrated to interact with melatonin receptors, and thus, we postulate that part of this system bias has its molecular basis in differences of the expression of receptor-associated proteins including heterodimerization partners. Finally, bias at the level of the receptor, by the expression of genetic receptor variants, will be discussed to show how a modified receptor function can have an effect on the risk for common diseases like type 2 diabetes in humans. LINKED ARTICLES: This article is part of a themed section on Recent Developments in Research of Melatonin and its Potential Therapeutic Applications. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.16/issuetoc.
Collapse
Affiliation(s)
- Erika Cecon
- Institut CochinInserm, U1016ParisFrance
- CNRS UMR 8104ParisFrance
- Univ. Paris Descartes, Sorbonne Paris CitéParisFrance
| | - Atsuro Oishi
- Institut CochinInserm, U1016ParisFrance
- CNRS UMR 8104ParisFrance
- Univ. Paris Descartes, Sorbonne Paris CitéParisFrance
| | - Ralf Jockers
- Institut CochinInserm, U1016ParisFrance
- CNRS UMR 8104ParisFrance
- Univ. Paris Descartes, Sorbonne Paris CitéParisFrance
| |
Collapse
|
32
|
Li Y, Li S, Zhou Y, Meng X, Zhang JJ, Xu DP, Li HB. Melatonin for the prevention and treatment of cancer. Oncotarget 2018; 8:39896-39921. [PMID: 28415828 PMCID: PMC5503661 DOI: 10.18632/oncotarget.16379] [Citation(s) in RCA: 247] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/09/2017] [Indexed: 12/17/2022] Open
Abstract
The epidemiological studies have indicated a possible oncostatic property of melatonin on different types of tumors. Besides, experimental studies have documented that melatonin could exert growth inhibition on some human tumor cells in vitro and in animal models. The underlying mechanisms include antioxidant activity, modulation of melatonin receptors MT1 and MT2, stimulation of apoptosis, regulation of pro-survival signaling and tumor metabolism, inhibition on angiogenesis, metastasis, and induction of epigenetic alteration. Melatonin could also be utilized as adjuvant of cancer therapies, through reinforcing the therapeutic effects and reducing the side effects of chemotherapies or radiation. Melatonin could be an excellent candidate for the prevention and treatment of several cancers, such as breast cancer, prostate cancer, gastric cancer and colorectal cancer. This review summarized the anticancer efficacy of melatonin, based on the results of epidemiological,experimental and clinical studies, and special attention was paid to the mechanisms of action.
Collapse
Affiliation(s)
- Ya Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Sha Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yue Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Xiao Meng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Jiao-Jiao Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Dong-Ping Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China.,South China Sea Bioresource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
33
|
Kurhaluk N, Bojková B, Winklewski PJ. Liver antioxidant and aerobic status improves after metformin and melatonin administration in a rat model of high-fat diet and mammary carcinogenesis. Can J Physiol Pharmacol 2018; 96:790-797. [PMID: 29658305 DOI: 10.1139/cjpp-2018-0001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Oxidative stress is involved in the development of various cancers. In the present study, the effect of long-term administration of peroral antidiabetic metformin and pineal hormone melatonin on liver antioxidant and aerobic status in female Sprague-Dawley rats carrying mammary tumors induced by N-methyl-N-nitrosourea was evaluated. Both substances were administered in a preventive and curative manner (12 days before and 16 weeks after the carcinogen application). Carcinogen administration induced oxidative stress: the level of thiobarbituric acid reactive substances (TBARS) considered as a marker of reactive oxygen species (ROS) generation in liver increased as well as the level of oxidatively modified protein content (OMP; aldehyde and ketone derivates). Metformin administration restored succinate dehydrogenase and lactate dehydrogenase activity and associated ROS production and OMP content to the level of intact rats, with predominant activation of superoxide dismutase (SOD) and glutathione reductase (GR). Melatonin alone and in combination with metformin also decreased TBARS content. OMP content decreased in all groups receiving chemoprevention. The rise in total antioxidant capacity after melatonin and particularly metformin and melatonin combination might result from the initiation of anaerobic metabolism and increasing SOD, GR, and glutathione peroxidase activity. Long-term administration of metformin and melatonin exerts antioxidant properties in liver, especially in combination.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- a Department of Zoology and Animal Physiology, Institute of Biology and Environment Protection, Pomeranian University, Slupsk, Poland
| | - Bianka Bojková
- b Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University, Košice, Slovak Republic
| | - Pawel J Winklewski
- c Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland.,d Department of Clinical Anatomy and Physiology, Faculty of Health Sciences, Pomeranian University of Slupsk, Slupsk, Poland
| |
Collapse
|
34
|
Shen YQ, Guerra-Librero A, Fernandez-Gil BI, Florido J, García-López S, Martinez-Ruiz L, Mendivil-Perez M, Soto-Mercado V, Acuña-Castroviejo D, Ortega-Arellano H, Carriel V, Diaz-Casado ME, Reiter RJ, Rusanova I, Nieto A, López LC, Escames G. Combination of melatonin and rapamycin for head and neck cancer therapy: Suppression of AKT/mTOR pathway activation, and activation of mitophagy and apoptosis via mitochondrial function regulation. J Pineal Res 2018; 64. [PMID: 29247557 DOI: 10.1111/jpi.12461] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) clearly involves activation of the Akt mammalian target of rapamycin (mTOR) signalling pathway. However, the effectiveness of treatment with the mTOR inhibitor rapamycin is often limited by chemoresistance. Melatonin suppresses neoplastic growth via different mechanisms in a variety of tumours. In this study, we aimed to elucidate the effects of melatonin on rapamycin-induced HNSCC cell death and to identify potential cross-talk pathways. We analysed the dose-dependent effects of melatonin in rapamycin-treated HNSCC cell lines (Cal-27 and SCC-9). These cells were treated with 0.1, 0.5 or 1 mmol/L melatonin combined with 20 nM rapamycin. We further examined the potential synergistic effects of melatonin with rapamycin in Cal-27 xenograft mice. Relationships between inhibition of the mTOR pathway, reactive oxygen species (ROS), and apoptosis and mitophagy reportedly increased the cytotoxic effects of rapamycin in HNSCC. Our results demonstrated that combined treatment with rapamycin and melatonin blocked the negative feedback loop from the specific downstream effector of mTOR activation S6K1 to Akt signalling, which decreased cell viability, proliferation and clonogenic capacity. Interestingly, combined treatment with rapamycin and melatonin-induced changes in mitochondrial function, which were associated with increased ROS production, increasing apoptosis and mitophagy. This led to increase cell death and cellular differentiation. Our data further indicated that melatonin administration reduced rapamycin-associated toxicity to healthy cells. Overall, our findings suggested that melatonin could be used as an adjuvant agent with rapamycin, improving effectiveness while minimizing its side effects.
Collapse
Affiliation(s)
- Ying-Qiang Shen
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
| | - Ana Guerra-Librero
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
| | - Beatriz I Fernandez-Gil
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
| | - Javier Florido
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
| | - Sergio García-López
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
| | - Laura Martinez-Ruiz
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
| | - Miguel Mendivil-Perez
- Medical Research Institute, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Viviana Soto-Mercado
- Medical Research Institute, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Darío Acuña-Castroviejo
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain
- CIBERFES, Ibs.Granada, Hospital Campus de la Salud, Granada, Spain
| | - Hector Ortega-Arellano
- Medical Research Institute, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Victor Carriel
- Department of Histology, Faculty of Medicine, University of Granada, Granada, Spain
| | - María E Diaz-Casado
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX, USA
| | - Iryna Rusanova
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
- CIBERFES, Ibs.Granada, Hospital Campus de la Salud, Granada, Spain
| | - Ana Nieto
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
| | - Luis C López
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain
- CIBERFES, Ibs.Granada, Hospital Campus de la Salud, Granada, Spain
| | - Germaine Escames
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain
- CIBERFES, Ibs.Granada, Hospital Campus de la Salud, Granada, Spain
| |
Collapse
|
35
|
Talib WH. Melatonin and Cancer Hallmarks. Molecules 2018; 23:molecules23030518. [PMID: 29495398 PMCID: PMC6017729 DOI: 10.3390/molecules23030518] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/09/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023] Open
Abstract
Melatonin is a natural indoleamine produced by the pineal gland that has many functions, including regulation of the circadian rhythm. Many studies have reported the anticancer effect of melatonin against a myriad of cancer types. Cancer hallmarks include sustained proliferation, evading growth suppressors, metastasis, replicative immortality, angiogenesis, resisting cell death, altered cellular energetics, and immune evasion. Melatonin anticancer activity is mediated by interfering with various cancer hallmarks. This review summarizes the anticancer role of melatonin in each cancer hallmark. The studies discussed in this review should serve as a solid foundation for researchers and physicians to support basic and clinical studies on melatonin as a promising anticancer agent.
Collapse
Affiliation(s)
- Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931-166, Jordan.
| |
Collapse
|
36
|
Melatonin and breast cancer: Evidences from preclinical and human studies. Crit Rev Oncol Hematol 2018; 122:133-143. [DOI: 10.1016/j.critrevonc.2017.12.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 10/20/2017] [Accepted: 12/27/2017] [Indexed: 12/22/2022] Open
|
37
|
de Almeida Chuffa LG, de Moura Ferreira G, Lupi LA, da Silva Nunes I, Fávaro WJ. P-MAPA immunotherapy potentiates the effect of cisplatin on serous ovarian carcinoma through targeting TLR4 signaling. J Ovarian Res 2018; 11:8. [PMID: 29343281 PMCID: PMC5773141 DOI: 10.1186/s13048-018-0380-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 01/11/2018] [Indexed: 01/16/2023] Open
Abstract
Background Toll-like receptors (TLRs) are transmembrane proteins expressed on the surface of ovarian cancer (OC) and immune cells. Identifying the specific roles of the TLR-mediated signaling pathways in OC cells is important to guide new treatments. Because immunotherapies have emerged as the adjuvant treatment for patients with OC, we investigated the effect of a promising immunotherapeutic strategy based on protein aggregate magnesium-ammonium phospholinoleate-palmitoleate anhydride (P-MAPA) combined with cisplatin (CIS) on the TLR2 and TLR4 signaling pathways via myeloid differentiation factor 88 (MyD88) and TLR-associated activator of interferon (TRIF) in an in vivo model of OC. Methods Tumors were chemically induced by a single injection of 100 μg of 7,12-dimethylbenz(a)anthracene (DMBA) directly under the left ovarian bursa in Fischer 344 rats. After the rats developed serous papillary OC, they were given P-MAPA, CIS or the combination P-MAPA+CIS as therapies. To understand the effects of the treatments, we assessed the tumor size, histopathology, and the TLR2- and TLR4-mediated inflammatory responses. Results Although CIS therapy was more effective than P-MAPA in reducing the tumor size, P-MAPA immunotherapy significantly increased the expressions of TLR2 and TLR4. More importantly, the combination of P-MAPA with CIS showed a greater survival rate compared to CIS alone, and exhibited a significant reduction in tumor volume compared to P-MAPA alone. The combination therapy also promoted the increase in the levels of the following OC-related proteins: TLR4, MyD88, TRIF, inhibitor of phosphorylated NF-kB alpha (p-IkBα), and nuclear factor kappa B (NF-kB p65) in both cytoplasmic and nuclear sites. While P-MAPA had no apparent effect on tumor necrosis factor alpha (TNF-α) and interleukin (IL)-6, it seems to increase interferon-γ (IFN-γ), which may induce the Thelper (Th1)-mediated immune response. Conclusion Collectively, our results suggest that P-MAPA immunotherapy combined with cisplatin could be considered an important therapeutic strategy against OC cells based on signaling pathways activated by TLR4.
Collapse
Affiliation(s)
- Luiz Gustavo de Almeida Chuffa
- Department of Anatomy, São Paulo State University (Unesp), Institute of Biosciences, Rubião Júnior, s/n, P.O Box: 18618-970, Botucatu, SP, 510, Brazil.
| | - Grazielle de Moura Ferreira
- Department of Anatomy, São Paulo State University (Unesp), Institute of Biosciences, Rubião Júnior, s/n, P.O Box: 18618-970, Botucatu, SP, 510, Brazil
| | - Luiz Antonio Lupi
- Department of Anatomy, São Paulo State University (Unesp), Institute of Biosciences, Rubião Júnior, s/n, P.O Box: 18618-970, Botucatu, SP, 510, Brazil
| | | | - Wagner José Fávaro
- Farmabrasilis R&D Division, Campinas, SP, Brazil.,Department of Structural and Functional Biology, Laboratory of Urogenital Carcinogenesis and Immunotherapy, UNICAMP - University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
38
|
Dehdashtian E, Mehrzadi S, Yousefi B, Hosseinzadeh A, Reiter RJ, Safa M, Ghaznavi H, Naseripour M. Diabetic retinopathy pathogenesis and the ameliorating effects of melatonin; involvement of autophagy, inflammation and oxidative stress. Life Sci 2018; 193:20-33. [DOI: 10.1016/j.lfs.2017.12.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/19/2017] [Accepted: 12/01/2017] [Indexed: 12/12/2022]
|
39
|
Lu H, Wu B, Ma G, Zheng D, Song R, Huang E, Mao M, Lu B. Melatonin represses oral squamous cell carcinoma metastasis by inhibiting tumor-associated neutrophils. Am J Transl Res 2017; 9:5361-5374. [PMID: 29312489 PMCID: PMC5752887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/08/2017] [Indexed: 06/07/2023]
Abstract
Tumor-associated neutrophils (TANs) promote metastasis of multiple cancers, including oral squamous cell carcinoma (OSCC). Melatonin (Mel) reportedly exerts anti-metastatic effects on OSCC. However, little is known about the anti-OSCC effects of Mel involved in TANs. In this study, intensive infiltration of TANs was positively associated with advanced stage, lymphatic metastasis, and poor prognosis of OSCC. Moreover, Mel reduced the survival and migration of OSCC-associated neutrophils. Mechanistically, Mel suppressed the TAN release of C-X-C motif chemokine ligand 8, C-C motif chemokine ligand 2 (CCL2), CCL4, and matrix metalloproteinase-9 by blockage of p38 MAPK and Akt signaling. Mel-fostered TANs decreased the migration and invasion of OSCC cells and reduced tube formation in vitro. Additionally, Mel-hampered pro-motility and pro-angiogenesis effects of TANs were dependent on MMP-9 suppression in OSCC. Overall, The beneficial roles of melatonin in retarding OSCC metastasis were implicated with inhibition of TANs.
Collapse
Affiliation(s)
- Haibin Lu
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital Affiliated to Dalian UniversityDalian, 116001 Liaoning, China
| | - Baolei Wu
- Department of Oral and Maxillofacial Surgery, No.3 Hospital of People’s Liberation ArmyBaoji 721004, Shannxi, China
| | - Ge Ma
- Department of Oral and Maxillofacial Surgery, No.3 Hospital of People’s Liberation ArmyBaoji 721004, Shannxi, China
| | - Deyu Zheng
- Department of Oral and Maxillofacial Surgery, No.3 Hospital of People’s Liberation ArmyBaoji 721004, Shannxi, China
| | - Ruijuan Song
- Department of Oral and Maxillofacial Surgery, No.3 Hospital of People’s Liberation ArmyBaoji 721004, Shannxi, China
| | - Erjiang Huang
- Department of Oral and Maxillofacial Surgery, No.3 Hospital of People’s Liberation ArmyBaoji 721004, Shannxi, China
| | - Ming Mao
- Department of Oral and Maxillofacial Surgery, No.3 Hospital of People’s Liberation ArmyBaoji 721004, Shannxi, China
| | - Bin Lu
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical UniversityXi’an 710032, Shannxi, China
| |
Collapse
|
40
|
Chuffa LGDA, Reiter RJ, Lupi LA. Melatonin as a promising agent to treat ovarian cancer: molecular mechanisms. Carcinogenesis 2017; 38:945-952. [PMID: 28575150 DOI: 10.1093/carcin/bgx054] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 06/01/2017] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer (OC) has the highest mortality rate of all gynecological cancers, and most patients develop chemoresistance after first-line treatments. Despite recent advances, the 5-year relative survival is ~45% for all OC subtypes, and invasive epithelial OC has only a 17% survival rate when diagnosed at a late stage. Identification of new efficacious molecules or biomarkers represents important opportunities in the treatment of OC. The pharmacological and physiological properties of melatonin indicate this agent could be useful against OC progression and metastasis. In normal cells, melatonin has potent antioxidant and anti-apoptotic actions. Conversely, melatonin has pro-oxidant as well as anti-proliferative, anti-angiogenic and immunomodulatory properties in many cancer types including hormone-dependent cancers. Although melatonin receptors have been identified in OC cells, the exact mechanism by which melatonin induces anticancer activities remains incompletely understood. Clinical studies have reported negative correlation between aggressiveness of OC and serum levels of melatonin, reinforcing the idea that melatonin may be a critical factor determining OC development. In vitro and in vivo studies suggest melatonin differentially regulates multiple signaling pathways in OC cells. This focused review explores the potential mechanisms of action of melatonin on cultured OC cells and in experimental models of OC in an attempt to clarify how melatonin modulates the signaling pathways involved in cancer cell apoptosis, survival, inflammation, proliferation and metabolic processes. Based on the evidence presented, we feel that melatonin, as an agent that controls cellular signals associated with malignancy, may be beneficial in combination with other therapeutics for OC treatment.
Collapse
Affiliation(s)
- Luiz Gustavo de Almeida Chuffa
- Department of Anatomy, Institute of Biosciences, UNESP - Universidade Estadual Paulista, Botucatu-SP, Brazil and Department of Cellular and Structural Biology, UTHSCSA, San Antonio, TX 78229, USA
| | - Russel J Reiter
- Department of Anatomy, Institute of Biosciences, UNESP - Universidade Estadual Paulista, Botucatu-SP, Brazil and Department of Cellular and Structural Biology, UTHSCSA, San Antonio, TX 78229, USA
| | - Luiz Antonio Lupi
- Department of Anatomy, Institute of Biosciences, UNESP - Universidade Estadual Paulista, Botucatu-SP, Brazil and Department of Cellular and Structural Biology, UTHSCSA, San Antonio, TX 78229, USA
| |
Collapse
|
41
|
Tsai TH, Lin CJ, Chua S, Chung SY, Yang CH, Tong MS, Hang CL. Melatonin attenuated the brain damage and cognitive impairment partially through MT2 melatonin receptor in mice with chronic cerebral hypoperfusion. Oncotarget 2017; 8:74320-74330. [PMID: 29088788 PMCID: PMC5650343 DOI: 10.18632/oncotarget.20382] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/19/2017] [Indexed: 12/13/2022] Open
Abstract
Background Vascular cognitive impairment (VCI) is a spectrum of cognitive impairment caused by various chronic diseases including aging, hypertension, and diabetes mellitus. Oxidative and inflammatory reactions induced by chronic cerebral hypoperfusion (CHP) are believed to cause VCI. Melatonin is reported to possess anti-oxidation and anti-inflammation effects. This study was designed to investigate the effect and mechanisms of melatonin in CHP mice model. Results The behavioral function results revealed that CHP mice were significantly impaired when compared with the control. Melatonin improved the cognitive function, but the addition of MT2 receptor antagonist reversed the improvement. The IHC staining showed melatonin significantly improved WM lesions and gliosis in CHP mice. Again, the addition of MT2 receptor antagonist to melatonin worsened the WM lesion and gliosis. Similar results were also found for mRNA and protein expressions of oxidative reaction and inflammatory cytokines. Materials and Method Forty C57BL/6 mice were divided into four groups: Group 1: sham control; Group 2: CHP mice; Group 3: CHP with melatonin treatment; Group 4: CHP-melatonin and MT2 receptor antagonist (all groups n = 10). Working memory was assessed with Y–arm test at day-28 post-BCAS (bilateral carotid artery stenosis). All mice were sacrificed at day-30 post-BCAS. The immunohistochemical (IHC) staining was used for white matter (WM) damage and gliosis. The expression of mRNA and proteins about inflammatory and oxidative reaction were measured and compared between groups. Conclusions Partially through MT2 receptor, melatonin is effective for CHP-induced brain damage.
Collapse
Affiliation(s)
- Tzu-Hsien Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheng-Jei Lin
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Sarah Chua
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Sheng-Ying Chung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Cheng-Hsu Yang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Meng-Shen Tong
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chi-Ling Hang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
42
|
Reiter RJ, Rosales-Corral SA, Tan DX, Acuna-Castroviejo D, Qin L, Yang SF, Xu K. Melatonin, a Full Service Anti-Cancer Agent: Inhibition of Initiation, Progression and Metastasis. Int J Mol Sci 2017; 18:E843. [PMID: 28420185 PMCID: PMC5412427 DOI: 10.3390/ijms18040843] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022] Open
Abstract
There is highly credible evidence that melatonin mitigates cancer at the initiation, progression and metastasis phases. In many cases, the molecular mechanisms underpinning these inhibitory actions have been proposed. What is rather perplexing, however, is the large number of processes by which melatonin reportedly restrains cancer development and growth. These diverse actions suggest that what is being observed are merely epiphenomena of an underlying more fundamental action of melatonin that remains to be disclosed. Some of the arresting actions of melatonin on cancer are clearly membrane receptor-mediated while others are membrane receptor-independent and involve direct intracellular actions of this ubiquitously-distributed molecule. While the emphasis of melatonin/cancer research has been on the role of the indoleamine in restraining breast cancer, this is changing quickly with many cancer types having been shown to be susceptible to inhibition by melatonin. There are several facets of this research which could have immediate applications at the clinical level. Many studies have shown that melatonin's co-administration improves the sensitivity of cancers to inhibition by conventional drugs. Even more important are the findings that melatonin renders cancers previously totally resistant to treatment sensitive to these same therapies. Melatonin also inhibits molecular processes associated with metastasis by limiting the entrance of cancer cells into the vascular system and preventing them from establishing secondary growths at distant sites. This is of particular importance since cancer metastasis often significantly contributes to death of the patient. Another area that deserves additional consideration is related to the capacity of melatonin in reducing the toxic consequences of anti-cancer drugs while increasing their efficacy. Although this information has been available for more than a decade, it has not been adequately exploited at the clinical level. Even if the only beneficial actions of melatonin in cancer patients are its ability to attenuate acute and long-term drug toxicity, melatonin should be used to improve the physical wellbeing of the patients. The experimental findings, however, suggest that the advantages of using melatonin as a co-treatment with conventional cancer therapies would far exceed improvements in the wellbeing of the patients.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | - Sergio A Rosales-Corral
- Centro de Investigacion Biomedica de Occidente, Del Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico.
| | - Dun-Xian Tan
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | | | - Lilan Qin
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan, Medical University, Taichung 40201, Taiwan.
| | - Kexin Xu
- Department of Molecular Medicine, UT Health, San Antonio, TX 78229, USA.
| |
Collapse
|
43
|
Zonta YR, Martinez M, Camargo ICC, Domeniconi RF, Lupi Júnior LA, Pinheiro PFF, Reiter RJ, Martinez FE, Chuffa LGA. Melatonin Reduces Angiogenesis in Serous Papillary Ovarian Carcinoma of Ethanol-Preferring Rats. Int J Mol Sci 2017; 18:ijms18040763. [PMID: 28398226 PMCID: PMC5412347 DOI: 10.3390/ijms18040763] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/24/2017] [Accepted: 03/30/2017] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is a hallmark of ovarian cancer (OC); the ingrowth of blood vessels promotes rapid cell growth and the associated metastasis. Melatonin is a well-characterized indoleamine that possesses important anti-angiogenic properties in a set of aggressive solid tumors. Herein, we evaluated the role of melatonin therapy on the angiogenic signaling pathway in OC of an ethanol-preferring rat model that mimics the same pathophysiological conditions occurring in women. OC was chemically induced with a single injection of 7,12-dimethylbenz(a)anthracene (DMBA) under the ovarian bursa. After the rats developed serous papillary OC, half of the animals received intraperitoneal injections of melatonin (200 µg/100 g body weight/day) for 60 days. Melatonin-treated animals showed a significant reduction in OC size and microvessel density. Serum levels of melatonin were higher following therapy, and the expression of its receptor MT1 was significantly increased in OC-bearing rats, regardless of ethanol intake. TGFβ1, a transforming growth factor-beta1, was reduced only after melatonin treatment. Importantly, vascular endothelial growth factor (VEGF) was severely reduced after melatonin therapy in animals given or not given ethanol. Conversely, the levels of VEGF receptor 1 (VEGFR1) was diminished after ethanol consumption, regardless of melatonin therapy, and VEGFR2 was only reduced following melatonin. Hypoxia-inducible factor (HIF)-1α was augmented with ethanol consumption, and, notably, melatonin significantly reduced their levels. Collectively, our results suggest that melatonin attenuates angiogenesis in OC in an animal model of ethanol consumption; this provides a possible complementary therapeutic opportunity for concurrent OC chemotherapy.
Collapse
MESH Headings
- Alcohol Drinking/physiopathology
- Animals
- Antioxidants/administration & dosage
- Antioxidants/pharmacology
- Blotting, Western
- Cystadenocarcinoma, Papillary/blood supply
- Cystadenocarcinoma, Papillary/drug therapy
- Cystadenocarcinoma, Papillary/metabolism
- Cystadenocarcinoma, Serous/blood supply
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/metabolism
- Ethanol/administration & dosage
- Female
- Food Preferences
- Immunohistochemistry
- Injections, Intraperitoneal
- Melatonin/administration & dosage
- Melatonin/pharmacology
- Microscopy, Fluorescence
- Neovascularization, Pathologic/prevention & control
- Ovarian Neoplasms/blood supply
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Rats
- Receptor, Melatonin, MT1/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-1/metabolism
Collapse
Affiliation(s)
- Yohan Ricci Zonta
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Marcelo Martinez
- Department of Morphology and Pathology, Universidade Federal de São Carlos (UFSCar), São Carlos-SP 13565-905, Brazil.
| | - Isabel Cristina C Camargo
- Department of Biotechnology, School of Sciences, Humanities and Languages, São Paulo State University (UNESP), Assis-SP 19806-900, Brazil.
| | - Raquel F Domeniconi
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Luiz Antonio Lupi Júnior
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Patricia Fernanda F Pinheiro
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, USA.
| | - Francisco Eduardo Martinez
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Luiz Gustavo A Chuffa
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| |
Collapse
|
44
|
Xu LX, Lv Y, Li YH, Ding X, Wang Y, Han X, Liu MH, Sun B, Feng X. Melatonin alleviates brain and peripheral tissue edema in a neonatal rat model of hypoxic-ischemic brain damage: the involvement of edema related proteins. BMC Pediatr 2017; 17:90. [PMID: 28351378 PMCID: PMC5371222 DOI: 10.1186/s12887-017-0824-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/02/2017] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Previous studies have indicated edema may be involved in the pathophysiology following hypoxic-ischemic encephalopathy (HIE), and melatonin may exhibit neuro-protection against brain insults. However, little is known regarding the mechanisms that involve the protective effects of melatonin in the brain and peripheral tissues after HIE. The present study aimed to examine the effects of melatonin on multiple organs, and the expression of edema related proteins in a neonatal rat model of hypoxic-ischemic brain damage (HIBD). METHODS One hundred ninety-two neonatal rats were randomly divided into three subgroups that underwent a sham surgery or HIBD. After the HIBD or sham-injury, the rats received an intraperitoneal injection of melatonin or an equal volume vehicle, respectively. We investigated the effects of melatonin on brain, kidney, and colon edema via histological examination and the expression of edema related proteins, including AQP-4, ZO-1 and occludin, via qPCR and western blot. RESULTS Our data indicated (1) Melatonin reduced the histological injury in the brain and peripheral organs induced by HIBD as assessed via H-E staining and transmission electron microscopy. (2) Melatonin alleviated the HIBD-induced cerebral edema characterized by increased brain water content. (3) HIBD induced significant changes of edema related proteins, such as AQP-4, ZO-1 and occludin, and these changes were partially reversed by melatonin treatment. CONCLUSIONS These findings provide substantial evidence that melatonin treatment has protective effects on the brain and peripheral organs after HIBD, and the edema related proteins, AQP4, ZO-1, and occludin, may indirectly contribute tothe mechanism of the edema protection by melatonin.
Collapse
Affiliation(s)
- Li-Xiao Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215006, China
| | - Yuan Lv
- Department of Neonatology, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Yan-Hong Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215006, China
| | - Xin Ding
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215006, China
| | - Ying Wang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215006, China
| | - Xing Han
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215006, China
| | - Ming-Hua Liu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215006, China
| | - Bin Sun
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215006, China.
| | - Xing Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
45
|
Chuffa LGA, Lupi Júnior LA, Seiva FRF, Martinez M, Domeniconi RF, Pinheiro PFF, dos Santos LD, Martinez FE. Quantitative Proteomic Profiling Reveals That Diverse Metabolic Pathways Are Influenced by Melatonin in an in Vivo Model of Ovarian Carcinoma. J Proteome Res 2016; 15:3872-3882. [DOI: 10.1021/acs.jproteome.6b00713] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Luiz Gustavo A. Chuffa
- Department
of Anatomy, Institute of Biosciences, UNESP − Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Luiz Antonio Lupi Júnior
- Department
of Anatomy, Institute of Biosciences, UNESP − Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Fábio R. F. Seiva
- Institute
of Biology, North of Parana State University − UENP, CLM, Bandeirantes, Paraná, Brazil
| | - Marcelo Martinez
- Department
of Morphology and Pathology, UFSCar − Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Raquel F. Domeniconi
- Department
of Anatomy, Institute of Biosciences, UNESP − Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Patricia Fernanda F. Pinheiro
- Department
of Anatomy, Institute of Biosciences, UNESP − Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Lucilene D. dos Santos
- Center
for the Study of Venoms and Venomous Animals (CEVAP), UNESP - Univ Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Francisco Eduardo Martinez
- Department
of Anatomy, Institute of Biosciences, UNESP − Universidade Estadual Paulista, Botucatu, São Paulo, Brazil
| |
Collapse
|
46
|
Lin SH, Huang YN, Kao JH, Tien LT, Tsai RY, Wong CS. Melatonin reverses morphine tolerance by inhibiting microglia activation and HSP27 expression. Life Sci 2016; 152:38-43. [PMID: 27012766 DOI: 10.1016/j.lfs.2016.03.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 03/06/2016] [Accepted: 03/17/2016] [Indexed: 01/14/2023]
Abstract
AIMS Melatonin has been reported to attenuate opioid tolerance. In this study, we explored the possible mechanism of melatonin in diminishing morphine tolerance. MAIN METHODS Two intrathecal (i.t.) catheters were implanted in male Wistar rats for drug delivery. One was linked to a mini-osmotic pump for morphine or saline infusion. On the seventh day, 50μg of melatonin or vehicle was injected through the other catheter instantly after discontinuation of morphine or saline infusion; 3h later, 15μg of morphine or saline was injected. The antinociceptive response was then measured using the tail-flick test every 30min for 120min. KEY FINDINGS The results showed that chronic morphine infusion elicited antinociceptive tolerance and upregulated heat shock protein 27 (HSP27) expression in the dorsal horn of the rat spinal cord. Melatonin pretreatment partially restored morphine's antinociceptive effect in morphine-tolerant rats and reversed morphine-induced HSP27 upregulation. In addition, chronic morphine infusion induced microglial cell activation and was reversed by melatonin treatment. SIGNIFICANCE The present study provides evidence that melatonin, acting via inhibiting morphine-induced neuroinflammation, can be useful as a therapeutic adjuvant for patients under long-term opioid treatment for pain relief.
Collapse
Affiliation(s)
- Sheng-Hsiung Lin
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei City, Taiwan; Medical Service Office, Tri-Service General Hospital Songshang Branch, Taipei City, Taiwan
| | - Ya-Ni Huang
- Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan City, Taiwan
| | - Jen-Hsin Kao
- Department of Anesthesiology, Cathay General Hospital, Taipei City, Taiwan; Cathay Medical Research Institute, Cathay General Hospital, Taipei City, Taiwan
| | - Lu-Tai Tien
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Ru-Yin Tsai
- Department of Nursing, Da-Yeh University, Changhua City, Taiwan
| | - Chih-Shung Wong
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei City, Taiwan; Department of Anesthesiology, Cathay General Hospital, Taipei City, Taiwan; School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
47
|
Chuffa LGA, Alves MS, Martinez M, Camargo ICC, Pinheiro PFF, Domeniconi RF, Júnior LAL, Martinez FE. Apoptosis is triggered by melatonin in an in vivo model of ovarian carcinoma. Endocr Relat Cancer 2016; 23:65-76. [PMID: 26555801 DOI: 10.1530/erc-15-0463] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/09/2015] [Indexed: 12/20/2022]
Abstract
Apoptosis plays an important role in the treatment of cancer, and targeting apoptosis-related molecules in ovarian cancer (OC) is of great therapeutic value. Melatonin (Mel) is an indoleamine displaying several anti-cancer properties and has been reported to modulate apoptosis signaling in multiple tumor subtypes. We investigated OC and the role of Mel therapy on the pro-apoptotic (p53, BAX, caspase-3, and cleaved caspase-3) and anti-apoptotic (Bcl-2 and survivin) proteins in an ethanol (EtOH)-preferring rat model. To induce OC, the left ovary was injected directly with a single dose of 100 μg 7,12-dimethylbenz(a)anthracene dissolved in 10 μl of sesame oil under the bursa. Right ovaries were used as sham-surgery controls. After developing OC, half of the animals received i.p. injections of Mel (200 μg/100 g BW per day) for 60 days. Body weight gain, EtOH consumption, and energy intake were unaffected by the treatments. Interestingly, absolute and relative OC masses showed a significant reduction after Mel therapy, regardless of EtOH consumption. To accomplish OC-related apoptosis, we first observed that p53, BAX, caspase-3, and cleaved caspase-3 were downregulated in OC tissue while Bcl-2 and survivin were overexpressed. Notably, Mel therapy and EtOH intake promoted apoptosis along with the upregulation of p53, BAX, and cleaved caspase-3. Fragmentation of DNA observed by TUNEL-positive nuclei was also enhanced following Mel treatment. In addition, Bcl-2 was downregulated by the EtOH intake and lower survivin levels were observed after Mel therapy. Taken together, these results suggest that Mel induce apoptosis in OC cells of EtOH-preferring animals.
Collapse
Affiliation(s)
- Luiz Gustavo A Chuffa
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Michelly S Alves
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Marcelo Martinez
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Isabel Cristina C Camargo
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Patricia F F Pinheiro
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Raquel F Domeniconi
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Luiz Antonio L Júnior
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| | - Francisco Eduardo Martinez
- Department of AnatomyInstitute of Biosciences of Botucatu, UNESP - Universidade Estadual Paulista, PO Box 18618-970, Rubião Júnior, s/n, Botucatu, São Paulo 510, BrazilDepartment of Morphology and PathologyUFSCar - Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, BrazilDepartment of Biological SciencesFaculty of Sciences and Letters, UNESP - Universidade Estadual Paulista, Assis, São Paulo 19806-900, Brazil
| |
Collapse
|
48
|
Melatonin attenuates the TLR4-mediated inflammatory response through MyD88- and TRIF-dependent signaling pathways in an in vivo model of ovarian cancer. BMC Cancer 2015; 15:34. [PMID: 25655081 PMCID: PMC4322437 DOI: 10.1186/s12885-015-1032-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/20/2015] [Indexed: 12/31/2022] Open
Abstract
Background Toll-like receptors (TLRs) are effector molecules expressed on the surface of ovarian cancer (OC) cells, but the functions of the TLR2/TLR4 signaling pathways in these cells remain unclear. Melatonin (mel) acts as an anti-inflammatory factor and has been reported to modulate TLRs in some aggressive tumor cell types. Therefore, we investigated OC and the effect of long-term mel therapy on the signaling pathways mediated by TLR2 and TLR4 via myeloid differentiation factor 88 (MyD88) and toll-like receptor-associated activator of interferon (TRIF) in an ethanol-preferring rat model. Methods To induce OC, the left ovary of animals either consuming 10% (v/v) ethanol or not was injected directly under the bursa with a single dose of 100 μg of 7,12-dimethylbenz(a)anthracene (DMBA) dissolved in 10 μL of sesame oil. The right ovaries were used as sham-surgery controls. After developing OC, half of the animals received i.p. injections of mel (200 μg/100 g b.w./day) for 60 days. Results Although mel therapy was unable to reduce TLR2 levels, it was able to suppress the OC-associated increase in the levels of the following proteins: TLR4, MyD88, nuclear factor kappa B (NFkB p65), inhibitor of NFkB alpha (IkBα), IkB kinase alpha (IKK-α), TNF receptor-associated factor 6 (TRAF6), TRIF, interferon regulatory factor 3 (IRF3), interferon β (IFN-β), tumor necrosis factor alpha (TNF-α), and interleukin (IL)-6. In addition, mel significantly attenuated the expression of IkBα, NFkB p65, TRIF and IRF-3, which are involved in TLR4-mediated signaling in OC during ethanol intake. Conclusion Collectively, our results suggest that mel attenuates the TLR4-induced MyD88- and TRIF-dependent signaling pathways in ethanol-preferring rats with OC.
Collapse
|