1
|
Nicotra R, Lutz C, Messal HA, Jonkers J. Rat Models of Hormone Receptor-Positive Breast Cancer. J Mammary Gland Biol Neoplasia 2024; 29:12. [PMID: 38913216 PMCID: PMC11196369 DOI: 10.1007/s10911-024-09566-0] [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: 03/29/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024] Open
Abstract
Hormone receptor-positive (HR+) breast cancer (BC) is the most common type of breast cancer among women worldwide, accounting for 70-80% of all invasive cases. Patients with HR+ BC are commonly treated with endocrine therapy, but intrinsic or acquired resistance is a frequent problem, making HR+ BC a focal point of intense research. Despite this, the malignancy still lacks adequate in vitro and in vivo models for the study of its initiation and progression as well as response and resistance to endocrine therapy. No mouse models that fully mimic the human disease are available, however rat mammary tumor models pose a promising alternative to overcome this limitation. Compared to mice, rats are more similar to humans in terms of mammary gland architecture, ductal origin of neoplastic lesions and hormone dependency status. Moreover, rats can develop spontaneous or induced mammary tumors that resemble human HR+ BC. To date, six different types of rat models of HR+ BC have been established. These include the spontaneous, carcinogen-induced, transplantation, hormone-induced, radiation-induced and genetically engineered rat mammary tumor models. Each model has distinct advantages, disadvantages and utility for studying HR+ BC. This review provides a comprehensive overview of all published models to date.
Collapse
Affiliation(s)
- Raquel Nicotra
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Amsterdam, Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| | - Hendrik A Messal
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| |
Collapse
|
2
|
Linowiecka K, Slominski AT, Reiter RJ, Böhm M, Steinbrink K, Paus R, Kleszczyński K. Melatonin: A Potential Regulator of DNA Methylation. Antioxidants (Basel) 2023; 12:1155. [PMID: 37371885 PMCID: PMC10295183 DOI: 10.3390/antiox12061155] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The pineal gland-derived indoleamine hormone, melatonin, regulates multiple cellular processes, ranging from chronobiology, proliferation, apoptosis, and oxidative damage to pigmentation, immune regulation, and mitochondrial metabolism. While melatonin is best known as a master regulator of the circadian rhythm, previous studies also have revealed connections between circadian cycle disruption and genomic instability, including epigenetic changes in the pattern of DNA methylation. For example, melatonin secretion is associated with differential circadian gene methylation in night shift workers and the regulation of genomic methylation during embryonic development, and there is accumulating evidence that melatonin can modify DNA methylation. Since the latter one impacts cancer initiation, and also, non-malignant diseases development, and that targeting DNA methylation has become a novel intervention target in clinical therapy, this review discusses the potential role of melatonin as an under-investigated candidate epigenetic regulator, namely by modulating DNA methylation via changes in mRNA and the protein expression of DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins. Furthermore, since melatonin may impact changes in the DNA methylation pattern, the authors of the review suggest its possible use in combination therapy with epigenetic drugs as a new anticancer strategy.
Collapse
Affiliation(s)
- Kinga Linowiecka
- Department of Human Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Andrzej T. Slominski
- Department of Dermatology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Pathology and Laboratory Medicine Service, VA Medical Center, Birmingham, AL 35294, USA
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA
| | - Markus Böhm
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Ralf Paus
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33125, USA
| | - Konrad Kleszczyński
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| |
Collapse
|
3
|
Gusman E, Standlee J, Reid KJ, Wolfe LF. Work-Related Sleep Disorders: Causes and Impacts. Semin Respir Crit Care Med 2023; 44:385-395. [PMID: 37072022 DOI: 10.1055/s-0043-1767787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Insufficient sleep syndrome, shift work disorder, and obstructive sleep apnea (OSA) not only significantly impact the health of affected individuals, but also pose a threat to public safety. This article describes the clinical manifestations and impact of these sleep disorders, particularly as they pertain to workers' health and those with safety-sensitive positions. Sleep deprivation, circadian rhythm disruptions, and excessive daytime sleepiness-hallmarks of insufficient sleep, shift work disorder, and OSA, respectively-all lead to a series of cognitive deficits and impaired concentration that affect workers in a wide variety of fields. We describe the health consequences of these disorders along with treatment strategies, with a focus on current regulatory standards and the under-recognition of OSA in commercial drivers. Given its large scale, there is a need for improved guidelines and regulations for the screening, diagnosis, treatment, and long-term follow-up of OSA in commercial motor vehicle drivers. Increased recognition of the ways in which these sleep disorders impact workers will pave the way for significant improvements in occupational health and safety.
Collapse
Affiliation(s)
- Elen Gusman
- Department of Pulmonary and Critical Care Medicine, Northwestern Medicine, Chicago, Illinois
| | - Jordan Standlee
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Kathryn J Reid
- Department of Neurology, Division of Sleep Medicine, Northwestern Medicine, Chicago, Illinois
- Center for Circadian and Sleep Medicine, Northwestern Medicine, Chicago, Illinois
| | - Lisa F Wolfe
- Department of Pulmonary and Critical Care Medicine, Northwestern Medicine, Chicago, Illinois
- Department of Neurology, Division of Sleep Medicine, Northwestern Medicine, Chicago, Illinois
- Center for Circadian and Sleep Medicine, Northwestern Medicine, Chicago, Illinois
| |
Collapse
|
4
|
Laborda-Illanes A, Sánchez-Alcoholado L, Castellano-Castillo D, Boutriq S, Plaza-Andrades I, Aranega-Martín L, Peralta-Linero J, Alba E, González-González A, Queipo-Ortuño MI. Development of in vitro and in vivo tools to evaluate the antiangiogenic potential of melatonin to neutralize the angiogenic effects of VEGF and breast cancer cells: CAM assay and 3D endothelial cell spheroids. Biomed Pharmacother 2023; 157:114041. [PMID: 36423543 DOI: 10.1016/j.biopha.2022.114041] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/15/2022] [Accepted: 11/19/2022] [Indexed: 11/22/2022] Open
Abstract
Melatonin is a molecule with different antitumor actions in breast cancer and has been described as an inhibitor of vascular endothelial growth factor (VEGF). Despite the recognition of the key role exerted by VEGF in tumor angiogenesis, limitations arise when developing models to test new antiangiogenic molecules. Thus, the aim of this study was to develop rapid, economic, high capacity and easy handling angiogenesis assays to test the antiangiogenic effects of melatonin and demonstrate its most effective dose to neutralize and interfere with the angiogenic sprouting effect induced by VEGF and MCF-7. To perform this, 3D endothelial cell (HUVEC) spheroids and a chicken embryo chorioallantoic membrane (CAM) assay were used. The results showed that VEGF and MCF-7 were able to stimulate the sprouting of the new vessels in 3D endothelial spheroids and the CAM assay, and that melatonin had an inhibitory effect on angiogenesis. Specifically, as the 1 mM pharmacological dose was the only effective dose able to inhibit the formation of ramifications around the alginate in the CAM assay model, this inhibition was shown to occur in a dose-dependent manner. Taken together, these techniques represent novel tools for the development of antiangiogenic molecules such as melatonin, with possible implications for the therapy of breast cancer.
Collapse
Affiliation(s)
- Aurora Laborda-Illanes
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain.
| | - Lidia Sánchez-Alcoholado
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain.
| | - Daniel Castellano-Castillo
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain.
| | - Soukaina Boutriq
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain.
| | - Isaac Plaza-Andrades
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain.
| | - Lucía Aranega-Martín
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain.
| | - Jesús Peralta-Linero
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain.
| | - Emilio Alba
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; Department of Medicine and Pediatrics. Faculty of Medicine, University of Malaga, 29071 Malaga, Spain.
| | - Alicia González-González
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; Department of Medicine and Pediatrics. Faculty of Medicine, University of Malaga, 29071 Malaga, Spain; Department of Physiology and Pharmacology. Faculty of Medicine, University of Cantabria, and Valdecilla Health Research Institute (IDIVAL), 39011 Santander, Spain.
| | - María Isabel Queipo-Ortuño
- Intercenter Medical Oncology Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga Biomedical Research Institute (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; Department of Surgical Specialties, Biochemical and Immunology. Faculty of Medicine, University of Málaga, 29071 Malaga, Spain.
| |
Collapse
|
5
|
Protective role of melatonin in breast cancer: what we can learn from women with blindness. Cancer Causes Control 2021; 33:1-13. [PMID: 34837156 DOI: 10.1007/s10552-021-01502-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/28/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE This review proposes an overall vision of the protective and therapeutic role of melatonin in breast cancer: from the specific cases of blind women and their reduction of breast cancer incidence to all clinical uses of the sleep hormone in breast cancer. METHODS We reviewed studies focused on (1) the correlation between blindness and breast cancer, (2) the correlation between melatonin and breast cancer occurrence in the general population, (3) melatonin therapeutic use in breast cancer, and (4) we discussed the properties of melatonin that could explain an anticancer effect. RESULTS (1) Seven studies of breast cancer risk in blind women related significant incidence decreases, up to 57%, among totally blind women. The limited number of studies and the absence of adjustment for confounding factors in most studies limit conclusions. None of these studies established melatonin profiles to determine whether blind women with a decreased breast cancer incidence produced higher levels of melatonin. (2) In the general population, 5 meta-analyses and 12 prospective-cohort studies focused on melatonin levels at recruitment and breast cancer occurrence. All reported the absence of correlation in premenopausal women, whereas in postmenopausal women, most studies showed significantly decreased risk for women with highest melatonin levels. (3) The therapeutic interest of melatonin associated with chemotherapy, radiotherapy, and hormonotherapy is poorly documented in breast cancer to conclude on a positive effect. (4) Melatonin effects on mammary carcinogenesis were only reported in in vitro and animal studies that demonstrated antiestrogenic, antioxidant, oncostatic, and immunomodulatory properties. CONCLUSION The preventive role of high endogenous melatonin on breast cancer as well as its beneficial therapeutic use remains to be proven.
Collapse
|
6
|
Fores-Martos J, Cervera-Vidal R, Sierra-Roca J, Lozano-Asencio C, Fedele V, Cornelissen S, Edvarsen H, Tadeo-Cervera I, Eroles P, Lluch A, Tabares-Seisdedos R, Falcó A, Van't Veer LJ, Schmidt M, Quigley DA, Børresen-Dale AL, Kristensen VN, Balmain A, Climent J. Circadian PERformance in breast cancer: a germline and somatic genetic study of PER3 VNTR polymorphisms and gene co-expression. NPJ Breast Cancer 2021; 7:118. [PMID: 34508103 PMCID: PMC8433453 DOI: 10.1038/s41523-021-00329-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/16/2021] [Indexed: 11/30/2022] Open
Abstract
Polymorphisms in the PER3 gene have been associated with several human disease phenotypes, including sleep disorders and cancer. In particular, the long allele of a variable number of tandem repeat (VNTR) polymorphism has been previously linked to an increased risk of breast cancer. Here we carried out a combined germline and somatic genetic analysis of the role of the PER3VNRT polymorphism in breast cancer. The combined data from 8284 individuals showed a non-significant trend towards increased breast cancer risk in the 5-repeat allele homozygous carriers (OR = 1.17, 95% CI: 0.97–1.42). We observed allelic imbalance at the PER3 locus in matched blood and tumor DNA samples, showing a significant retention of the long variant (risk) allele in tumor samples, and a preferential loss of the short repetition allele (p = 0.0005). Gene co-expression analysis in healthy and tumoral breast tissue samples uncovered significant associations between PER3 expression levels with those from genes which belong to several cancer-associated pathways. Finally, relapse-free survival (RFS) analysis showed that low expression levels of PER3 were linked to a significant lower RSF in luminal A (p = 3 × 10−12) but not in the rest of breast cancer subtypes.
Collapse
Affiliation(s)
- Jaume Fores-Martos
- ESI International Chair at CEU-UCH, CEU Universities, Valencia, Spain.,Biomedical Research Networking Center of Mental Health (CIBERSAM), Madrid, Spain
| | | | | | - Carlos Lozano-Asencio
- INCLIVA Research Institute. Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Vita Fedele
- Digestive Molecular Clinical Oncology Research Unit, Section of Medical Oncology, Department of Medicine, University of Verona, Verona, Italy
| | - Sten Cornelissen
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Hege Edvarsen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Irene Tadeo-Cervera
- Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos. Facultad de Veterinaria, Universidad CEU Cardenal Herrera. CEU Universities, Valencia, Spain
| | - Pilar Eroles
- INCLIVA Research Institute. Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Ana Lluch
- INCLIVA Research Institute. Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Rafa Tabares-Seisdedos
- Biomedical Research Networking Center of Mental Health (CIBERSAM), Madrid, Spain.,Department of Medicine, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Antonio Falcó
- ESI International Chair at CEU-UCH, CEU Universities, Valencia, Spain.,Departamento de Matemáticas, Física y Ciencias Tecnológicas, Escuela Superior de Enseñanzas Técnicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Laura J Van't Veer
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Marjanka Schmidt
- Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos. Facultad de Veterinaria, Universidad CEU Cardenal Herrera. CEU Universities, Valencia, Spain
| | - David A Quigley
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.,Departments of Urology and Epidemiology & Biostatistics, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Vessela N Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Allan Balmain
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Joan Climent
- ESI International Chair at CEU-UCH, CEU Universities, Valencia, Spain. .,INCLIVA Research Institute. Hospital Clínico Universitario de Valencia, Valencia, Spain. .,Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos. Facultad de Veterinaria, Universidad CEU Cardenal Herrera. CEU Universities, Valencia, Spain.
| |
Collapse
|
7
|
Immunomodulation: An immune regulatory mechanism in carcinoma therapeutics. Int Immunopharmacol 2021; 99:107984. [PMID: 34303999 DOI: 10.1016/j.intimp.2021.107984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/29/2021] [Accepted: 07/11/2021] [Indexed: 01/01/2023]
Abstract
Cancer has been generally related to the possession of numerous mutations which interrupt important signaling pathways. Nevertheless, deregulated immunological signaling is considered as one of the key factors associated with the development and progression of cancer. The signaling pathways operate as modular network with different components interacting in a switch-like fashion with two proteins interplaying between each other leading to direct or indirect inhibition or stimulation of down-stream factors. Genetic, epigenetic, and transcriptomic alterations maintain the pathological conduit of different signaling pathways via affecting diverse mechanisms including cell destiny. At present, immunotherapy is one of the best therapies opted for cancer treatment. The cancer immunotherapy strategy includes harnessing the specificity and killing mechanisms of the immunological system to target and eradicate malignant cells. Targeted therapies utilizing several little molecules including Galunisertib, Astragaloside-IV, Melatonin, and Jervine capable of regulating key signaling pathways can effectively help in the management of different carcinomas.
Collapse
|
8
|
Laborda-Illanes A, Sánchez-Alcoholado L, Boutriq S, Plaza-Andrades I, Peralta-Linero J, Alba E, González-González A, Queipo-Ortuño MI. A New Paradigm in the Relationship between Melatonin and Breast Cancer: Gut Microbiota Identified as a Potential Regulatory Agent. Cancers (Basel) 2021; 13:3141. [PMID: 34201776 PMCID: PMC8269379 DOI: 10.3390/cancers13133141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/18/2023] Open
Abstract
In this review we summarize a possible connection between gut microbiota, melatonin production, and breast cancer. An imbalance in gut bacterial population composition (dysbiosis), or changes in the production of melatonin (circadian disruption) alters estrogen levels. On the one hand, this may be due to the bacterial composition of estrobolome, since bacteria with β-glucuronidase activity favour estrogens in a deconjugated state, which may ultimately lead to pathologies, including breast cancer. On the other hand, it has been shown that these changes in intestinal microbiota stimulate the kynurenine pathway, moving tryptophan away from the melatonergic pathway, thereby reducing circulating melatonin levels. Due to the fact that melatonin has antiestrogenic properties, it affects active and inactive estrogen levels. These changes increase the risk of developing breast cancer. Additionally, melatonin stimulates the differentiation of preadipocytes into adipocytes, which have low estrogen levels due to the fact that adipocytes do not express aromatase. Consequently, melatonin also reduces the risk of breast cancer. However, more studies are needed to determine the relationship between microbiota, melatonin, and breast cancer, in addition to clinical trials to confirm the sensitizing effects of melatonin to chemotherapy and radiotherapy, and its ability to ameliorate or prevent the side effects of these therapies.
Collapse
Affiliation(s)
- Aurora Laborda-Illanes
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
| | - Lidia Sánchez-Alcoholado
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
| | - Soukaina Boutriq
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
| | - Isaac Plaza-Andrades
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
| | - Jesús Peralta-Linero
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
| | - Emilio Alba
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Centro de Investigación Biomédica en Red de Cáncer (Ciberonc CB16/12/00481), 28029 Madrid, Spain
| | - Alicia González-González
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
| | - María Isabel Queipo-Ortuño
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain; (A.L.-I.); (L.S.-A.); (S.B.); (I.P.-A.); (J.P.-L.); (M.I.Q.-O.)
- Centro de Investigación Biomédica en Red de Cáncer (Ciberonc CB16/12/00481), 28029 Madrid, Spain
| |
Collapse
|
9
|
Mehrzadi S, Pourhanifeh MH, Mirzaei A, Moradian F, Hosseinzadeh A. An updated review of mechanistic potentials of melatonin against cancer: pivotal roles in angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress. Cancer Cell Int 2021; 21:188. [PMID: 33789681 PMCID: PMC8011077 DOI: 10.1186/s12935-021-01892-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/23/2021] [Indexed: 12/19/2022] Open
Abstract
Cancers are serious life-threatening diseases which annually are responsible for millions of deaths across the world. Despite many developments in therapeutic approaches for affected individuals, the rate of morbidity and mortality is high. The survival rate and life quality of cancer patients is still low. In addition, the poor prognosis of patients and side effects of the present treatments underscores that finding novel and effective complementary and alternative therapies is a critical issue. Melatonin is a powerful anticancer agent and its efficiency has been widely documented up to now. Melatonin applies its anticancer abilities through affecting various mechanisms including angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress. Regarding the implication of mentioned cellular processes in cancer pathogenesis, we aimed to further evaluate the anticancer effects of melatonin via these mechanisms.
Collapse
Affiliation(s)
- Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mirzaei
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Farid Moradian
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Zheng Y, Jiang H, Wang HQ, Guo HX, Han DX, Huang YJ, Gao Y, Yuan B, Zhang JB. Identifying daily changes in circRNAs and circRNA-associated-ceRNA networks in the rat pineal gland. Int J Med Sci 2021; 18:1225-1239. [PMID: 33526984 PMCID: PMC7847611 DOI: 10.7150/ijms.51743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
Circular RNAs (circRNAs) are a new class of covalently closed circular RNA molecules that are involved in many biological processes. However, information about circRNAs in the pineal gland, particularly that of rats, is limited. To establish resources for the study of the rat pineal gland, we performed transcriptome analysis of the pineal glands during the day and night. In this study, 1413 circRNAs and 1989 miRNAs were identified in the pineal gland of rats during the night and day using the Illumina platform. Forty differentially expressed circRNAs and 93 differentially expressed miRNAs were obtained, among which 20 circRNAs and 37 miRNAs were significantly upregulated during the day and 20 circRNAs and 56 miRNAs were significantly upregulated during the night. As circRNAs have been reported to work as miRNA sponges, we predicted 15940 interactions among 40 circRNAs, 93 miRNAs and 400 mRNAs with differential diurnal expression using miRanda and TargetScan to build a ceRNA regulatory network in the rat pineal gland. The diurnal expression profile of circRNAs in the rat pineal gland may provide additional information about the role of circRNAs in regulating changes in melatonin circadian rhythms. The analyzed data reported in this study will be an important resource for future studies to elucidate the altered physiology of circRNAs in diurnal rhythms.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| | - Hao Jiang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| | - Hao-Qi Wang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| | - Hai-Xiang Guo
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| | - Dong-Xu Han
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| | - Yi-Jie Huang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| | - Yan Gao
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| | - Bao Yuan
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| | - Jia-Bao Zhang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, Jilin, P.R. China
| |
Collapse
|
11
|
Mehrafza M, Asgharnia M, Raoufi A, Hosseinzadeh E, Samadnia S, Roushan ZA. The effect of seasonality on reproductive outcome of patients undergoing intracytoplasmic sperm injection: A descriptive cross-sectional study. Int J Reprod Biomed 2020; 18:989-994. [PMID: 33349808 PMCID: PMC7749974 DOI: 10.18502/ijrm.v13i11.7967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 12/04/2019] [Accepted: 06/13/2020] [Indexed: 11/24/2022] Open
Abstract
Background There is conflicting evidence regarding the impact of season on the assisted reproductive technology outcome. Objective To retrospectively compare three year outcome of women undergoing their first intracytoplasmic sperm injection cycle, across seasons. Materials and Methods In this descriptive cross-sectional study, 3,670 women who underwent their first intracytoplasmic sperm injection cycle in Mehr Medical Institute, Rasht, Iran between April 2010 and May 2014 were studied. Women were divided into four groups according to the day of oocyte retrival as: spring (n = 808), summer (n = 994), autumn (n = 1066), and winter (n = 802). Basal and stimulation charecteristics were compared among groups. Results While sperm concentration and motility were significantly lower during summer, the total number of retrieved and metaphase II oocytes were significantly higher (p = 0.0001, p = 0.0001, p = 0.004, p = 0.02, respectively). Fertilization rate were significantly higher during autumn (p = 0.0001). Also, the number of high- quality transferred embryos were significantly higher during summer and winter (p = 0.03). A similar pattern was observed in implantation rate and pregnancy over the four seasons. Conclusion Despite the fact that intracytoplasmic sperm injection minimize the seasonal effect on pregnancy outcome, changes in pregnancy rate still occur among different seasons without particular pattern. It seems that performing assisted reproductive technology procedures in a particular season should be considered as an effective factor.
Collapse
Affiliation(s)
- Marzieh Mehrafza
- Mehr Fertility Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Maryam Asgharnia
- Mehr Fertility Research Center, Guilan University of Medical Sciences, Rasht, Iran.,Reproductive Health Research Center, Department of Obstetrics and Gynecology, Alzahra Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Azadeh Raoufi
- Mehr Fertility Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Elmira Hosseinzadeh
- Mehr Fertility Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Sajedeh Samadnia
- Mehr Fertility Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Atrkar Roushan
- Reproductive Health Research Center, Department of Obstetrics and Gynecology, Alzahra Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
12
|
Maroufi NF, Ashouri N, Mortezania Z, Ashoori Z, Vahedian V, Amirzadeh-Iranaq MT, Fattahi A, Kazemzadeh H, Bizzarri M, Akbarzadeh M, Nejabati HR, Faridvand Y, Rashidi MR, Nouri M. The potential therapeutic effects of melatonin on breast cancer: An invasion and metastasis inhibitor. Pathol Res Pract 2020; 216:153226. [PMID: 32987338 DOI: 10.1016/j.prp.2020.153226] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/30/2020] [Accepted: 09/11/2020] [Indexed: 01/14/2023]
Abstract
Breast cancer is the most common cancer among women and its metastasis which generally observed at the last stage is the major cause of breast cancer-related death. Therefore, the agents that have the potential to prevent metastatic and invasive nature of breast cancer can open up new therapeutic strategies. Melatonin, a major hormone of pineal gland, is a powerful anti-cancer agent. There are growing evidence regarding the protective effect of melatonin against cancer invasion and metastasis. The anti-metastatic feature of melatonin accompanies with suppression of tumor proliferation, induction of tumor apoptosis, regulation of the cell cycle, modulating angiogenesis, impediment of invasion, and induction of cancer cells sensitivity to the chemotherapy agents. More recently, anti-metastatic effect of melatonin through affecting cancer stem cells and vascular mimicry has been identified. Thus, the aim of this review is to discuss the potential therapeutic effect of melatonin on breast cancer via modulating the cells invasion and metastasis.
Collapse
Affiliation(s)
- Nazila Fathi Maroufi
- Student Research Committee, Tabriz University of Medical Sciences, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Ashouri
- Department of Medicine, Faculty of Cellular and Molecular Biology, University of Shiraz, Iran
| | | | - Zahra Ashoori
- Department of Medical, Faculty of Medical, University of Shahid Beheshti, Iran
| | - Vahid Vahedian
- Department of Experimental Biomedicine, Dr. Vahid Vahedian Medical Diagnostic Laboratory Gorgan, Iran; Department of Medical Laboratory Sciences, Faculty of Medicine, Islamic Azad University (IAU), Sari, Iran
| | | | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Kazemzadeh
- Department of Biology, Faculty of Basic Sciences, Hakim Sabzevari University, Iran
| | - Mariano Bizzarri
- University La Sapienza, Department of Experimental Medicine, System Biology Group, Rome, Italy
| | - Maryam Akbarzadeh
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Faridvand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Wen YC, Lin YW, Chu CY, Yang YC, Yang SF, Liu YF, Hsiao M, Lee WJ, Chien MH. Melatonin-triggered post-transcriptional and post-translational modifications of ADAMTS1 coordinately retard tumorigenesis and metastasis of renal cell carcinoma. J Pineal Res 2020; 69:e12668. [PMID: 32408377 DOI: 10.1111/jpi.12668] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/25/2020] [Accepted: 05/07/2020] [Indexed: 12/27/2022]
Abstract
A disintegrin and metalloprotease with thrombospondin motifs (ADAMTS) family are widely implicated in tissue remodeling events manifested in cancer development. ADAMTS1, the most fully characterized ADAMTS, plays conflicting roles in different cancer types; however, the role of ADAMTS1 in renal cell carcinoma (RCC) remains unclear. Herein, we found that ADAMTS1 is highly expressed in RCC tissues compared to normal renal tissues, and its expression was correlated with an advanced stage and a poor prognosis of RCC patients. In vitro, we observed higher expression of ADAMTS1 in metastatic (m)RCC cells compared to primary cells, and manipulation of ADAMTS1 expression affected cell invasion and clonogenicity. Results from protease array showed that ADAMTS1 is modulated by melatonin through mechanisms independent of the MT1 receptor in mRCC cells, and overexpression of ADAMTS1 relieved the invasion/clonogenicity and growth/metastasis inhibition imposed by melatonin treatment in vitro and in an orthotopic xenograft model. The human microRNA (miR) OneArray showed that miR-181d and miR-let-7f were induced by melatonin and, respectively, targeted the 3'-UTR and non-3'-UTR of ADAMTS1 to suppress its expression and mRCC invasive ability. Clinically, RCC patients with high levels of miR-181d or miR-let-7f and a low level of ADAMTS1 had the most favorable prognoses. In addition, ubiquitin/proteasome-mediated degradation of ADAMTS1 can also be triggered by melatonin. Together, our study indicates that ADAMTS1 may be a useful biomarker for predicting RCC progression. The novel convergence between melatonin and ADAMTS1 post-transcriptional and post-translational regulation provides new insights into the role of melatonin-induced molecular regulation in suppressing RCC progression.
Collapse
Affiliation(s)
- Yu-Ching Wen
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Yung-Wei Lin
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Chih-Ying Chu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Fan Liu
- Department of Biomedical Sciences, College of Medicine Sciences and Technology, Chung Shan Medical University, Taichung, Taiwan
- Division of Allergy, Department of Pediatrics, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital Taipei, Taiwan
| |
Collapse
|
14
|
Alonso-González C, González A, Menéndez-Menéndez J, Martínez-Campa C, Cos S. Melatonin as a Radio-Sensitizer in Cancer. Biomedicines 2020; 8:E247. [PMID: 32726912 PMCID: PMC7460067 DOI: 10.3390/biomedicines8080247] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is one of the treatments of choice in many types of cancer. Adjuvant treatments to radiotherapy try, on one hand, to enhance the response of tumor cells to radiation and, on the other hand, to reduce the side effects to normal cells. Radiosensitizers are agents that increase the effect of radiation in tumor cells by trying not to increase side effects in normal tissues. Melatonin is a hormone produced mainly by the pineal gland which has an important role in the regulation of cancer growth, especially in hormone-dependent mammary tumors. Different studies have showed that melatonin administered with radiotherapy is able to enhance its therapeutic effects and can protect normal cells against side effects of this treatment. Several mechanisms are involved in the radiosensitization induced by melatonin: increase of reactive oxygen species production, modulation of proteins involved in estrogen biosynthesis, impairment of tumor cells to DNA repair, modulation of angiogenesis, abolition of inflammation, induction of apoptosis, stimulation of preadipocytes differentiation and modulation of metabolism. At this moment, there are very few clinical trials that study the therapeutic usefulness to associate melatonin and radiotherapy in humans. All findings point to melatonin as an effective adjuvant molecule to radiotherapy in cancer treatment.
Collapse
Affiliation(s)
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (J.M.-M.); (S.C.)
| | | | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain; (C.A.-G.); (J.M.-M.); (S.C.)
| | | |
Collapse
|
15
|
Hasan M, Browne E, Guarinoni L, Darveau T, Hilton K, Witt-Enderby PA. Novel Melatonin, Estrogen, and Progesterone Hormone Therapy Demonstrates Anti-Cancer Actions in MCF-7 and MDA-MB-231 Breast Cancer Cells. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2020; 14:1178223420924634. [PMID: 32636633 PMCID: PMC7318814 DOI: 10.1177/1178223420924634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/13/2020] [Indexed: 12/28/2022]
Abstract
A novel melatonin, estrogen, and progesterone hormone therapy was developed as a safe bio-identical alternative hormone therapy for menopausal women based on the Women’s Health Initiative findings that PremPro™ increased breast cancer risk and mortality of all types of breast cancer in postmenopausal women. For HER2 breast cancer, melatonin, estrogen, and progesterone delayed tumor onset and reduced tumor incidence in neu female mice. For other breast cancers, its actions are unknown. In this study, melatonin, estrogen, and progesterone hormone therapy were assessed in human ER+ (MCF-7) and triple negative breast cancer (MDA-MB-231) cells, and found to decrease proliferation and migration of both breast cancer lines. Inhibition of MEK1/2 and 5 using PD98059 and BIX02189, respectively, inhibited proliferation and migration in MDA-MB-231 cells and proliferation in MCF-7 cells; however, when combined with melatonin, estrogen, and progesterone, BIX02189 blocked melatonin, estrogen, and progesterone–mediated inhibition of migration in MCF-7 cells and induced Elf-5. For MDA-MB-231 cells, BIX02189 combined with melatonin, estrogen, and progesterone inhibited proliferation and increased pERK1/2 and β1-INTEGRIN; levels of pERK5 remained low/nearly absent in both breast cancer lines. These findings demonstrate novel anti-cancer actions of melatonin, estrogen, and progesterone in ER+ and triple negative breast cancer cells through intricate MEK1/2- and MEK5-associated signaling cascades that favor anti-proliferation and anti-migration.
Collapse
Affiliation(s)
- Mahmud Hasan
- Division of Pharmaceutical, Administrative and Social Sciences, Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Erin Browne
- Division of Pharmaceutical, Administrative and Social Sciences, Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Laura Guarinoni
- Division of Pharmaceutical, Administrative and Social Sciences, Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Travis Darveau
- Division of Pharmaceutical, Administrative and Social Sciences, Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Katherine Hilton
- Division of Pharmaceutical, Administrative and Social Sciences, Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Paula A Witt-Enderby
- Division of Pharmaceutical, Administrative and Social Sciences, Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
16
|
González-González A, González A, Rueda N, Alonso-González C, Menéndez JM, Martínez-Campa C, Mitola S, Cos S. Usefulness of melatonin as complementary to chemotherapeutic agents at different stages of the angiogenic process. Sci Rep 2020; 10:4790. [PMID: 32179814 PMCID: PMC7076026 DOI: 10.1038/s41598-020-61622-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 02/22/2020] [Indexed: 02/07/2023] Open
Abstract
Chemotherapeutics are sometimes administered with drugs, like antiangiogenic compounds, to increase their effectiveness. Melatonin exerts antitumoral actions through antiangiogenic actions. We studied if melatonin regulates the response of HUVECs to chemotherapeutics (docetaxel and vinorelbine). The inhibition that these agents exert on some of the processes involved in angiogenesis, such as, cell proliferation, migratory capacity or vessel formation, was enhanced by melatonin. Regarding to estrogen biosynthesis, melatonin impeded the negative effect of vinorelbine, by decreasing the activity and expression of aromatase and sulfatase. Docetaxel and vinorelbine increased the expression of VEGF-A, VEGF-B, VEGF-C, VEGFR-1, VEGFR-3, ANG1 and/or ANG-2 and melatonin inhibited these actions. Besides, melatonin prevented the positive actions that docetaxel exerts on the expression of other factors related to angiogenesis like JAG1, ANPEP, IGF-1, CXCL6, AKT1, ERK1, ERK2, MMP14 and NOS3 and neutralized the stimulating actions of vinorelbine on the expression of FIGF, FGFR3, CXCL6, CCL2, ERK1, ERK2, AKT1, NOS3 and MMP14. In CAM assay melatonin inhibited new vascularization in combination with chemotherapeutics. Melatonin further enhanced the chemotherapeutics-induced inhibition of p-AKT and p-ERK and neutralized the chemotherapeutics-caused stimulatory effect on HUVECs permeability by modifying the distribution of VE cadherin. Our results confirm that melatonin blocks proangiogenic and potentiates antiangiogenic effects induced by docetaxel and vinorelbine enhancing their antitumor effectiveness.
Collapse
Affiliation(s)
- Alicia González-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011, Santander, Spain.
| | - Noemi Rueda
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Javier Menéndez Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011, Santander, Spain.
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, Laboratory for Preventive and Personalized Medicine, University of Brescia, 25123, Brescia, Italy
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011, Santander, Spain
| |
Collapse
|
17
|
Hsieh MJ, Lin CW, Su SC, Reiter RJ, Chen AWG, Chen MK, Yang SF. Effects of miR-34b/miR-892a Upregulation and Inhibition of ABCB1/ABCB4 on Melatonin-Induced Apoptosis in VCR-Resistant Oral Cancer Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:877-889. [PMID: 31982774 PMCID: PMC6994412 DOI: 10.1016/j.omtn.2019.12.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 11/27/2019] [Accepted: 12/19/2019] [Indexed: 12/19/2022]
Abstract
Multidrug resistance (MDR) is the resistance of cells toward various drugs commonly used in tumor treatment. The mechanism of drug resistance in oral cancer is not completely understood. Melatonin is an endogenously produced molecule involved in active biological mechanisms including antiproliferation, oncogene expression modulation, antitumor invasion and migration, and anti-inflammatory, antioxidant, and antiangiogenic effects. Despite these functions, the effects of melatonin on vincristine (VCR)-resistant human oral cancer cells remain largely unknown. This study analyzed the role of melatonin in VCR-resistant human oral cancer cells along with the underlying mechanism. We determined that melatonin induced the apoptosis and autophagy of VCR-resistant oral cancer cells; these actions were mediated by AKT, p38, and c-Jun N-terminal kinase (JNK). Melatonin inhibited ATP-binding cassette B1 (ABCB1) and ABCB4 expression in vitro and in vivo. Melatonin reduced the drug resistance and promoted the apoptosis of VCR-resistant oral cancer cells through the upregulation of microRNA-892a (miR-892a) and miR-34b-5p expressions. The expression of miR-892a and miR-34b-5p was related to melatonin-induced apoptosis, but not autophagy. Therefore, melatonin is a potential novel chemotherapeutic agent for VCR-resistant human oral cancer cell lines.
Collapse
Affiliation(s)
- Ming-Ju Hsieh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 402, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital 402, Taichung, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung 204, Taiwan; Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Linkou, and Keelung 204, Taiwan
| | - Russel J Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Andy Wei-Ge Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Mu-Kuan Chen
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan.
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
18
|
Fathizadeh H, Mirzaei H, Asemi Z. Melatonin: an anti-tumor agent for osteosarcoma. Cancer Cell Int 2019; 19:319. [PMID: 31798348 PMCID: PMC6884844 DOI: 10.1186/s12935-019-1044-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/22/2019] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma is the most common bone tumors which consisted of malignant mesenchymal cells generating osteoid and immature bone. It has been showed that osteosarcoma is common in children and adolescents and shows high mortality rate. A variety of therapeutic approaches (i.e., resection surgery, combined with chemotherapy and radiotherapy) have been used as conventional treatments in patients with osteosarcoma. Despite several attempts to improve therapeutic response, the rate of survival for osteosarcoma has not changed during the past 3 decades. Therefore, the discovery and developing new effective therapeutic platforms are required. Along to the established anti-cancer agents, some physiological regulators such melatonin, have been emerged as new anti-cancer agents. Melatonin is an indolamine hormone which is secreted from the pineal glands during the night and acts as physiological regulator. Given that melatonin shows a wide spectrum anti-tumor impacts. Besides different biologic activities of melatonin (e.g., immunomodulation and antioxidant properties), melatonin has a crucial role in the formation of bones, and its deficiency could be directly related to bone cancers. Several in vitro and in vivo experiments evaluated the effects of melatonin on osteosarcoma and other types of bone cancer. Taken together, the results of these studies indicated that melatonin could be introduced as new therapeutic candidate or as adjuvant in combination with other anti-tumor agents in the treatment of osteosarcoma. Herein, we summarized the anti-tumor effects of melatonin for osteosarcoma cancer as well as its mechanism of action.
Collapse
Affiliation(s)
- Hadis Fathizadeh
- 1Department of Microbiology, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Hamed Mirzaei
- 2Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zatollah Asemi
- 2Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| |
Collapse
|
19
|
Agbaria S, Haim A, Fares F, Zubidat AE. Epigenetic modification in 4T1 mouse breast cancer model by artificial light at night and melatonin - the role of DNA-methyltransferase. Chronobiol Int 2019; 36:629-643. [PMID: 30746962 DOI: 10.1080/07420528.2019.1574265] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Currently, one of the most disputed hypotheses regarding breast cancer (BC) development is exposure to short wavelength artificial light at night (ALAN) as multiple studies suggest a possible link between them. This link is suggested to be mediated by nocturnal melatonin suppression that plays an integral role in circadian regulations including cell division. The objective of the research was to evaluate effects of 1 × 30 min/midnight ALAN (134 µ Wcm-2, 460 nm) with or without nocturnal melatonin supplement on tumor development and epigenetic responses in 4T1 tumor-bearing BALB/c mice. Mice were monitored for body mass (Wb) and tumor volume for 3 weeks and thereafter urine samples were collected at regular intervals for determining daily rhythms of 6-sulfatoxymelatonin (6-SMT). Finally, mice were sacrificed and the tumor, lungs, liver, and spleen were excised for analyzing the total activity of DNA methyltransferases (DNMT) and global DNA methylation (GDM) levels. Mice exposed to ALAN significantly reduced 6-SMT levels and increased Wb, tumor volume, and lung metastasis compared with controls. These effects were diminished by melatonin. The DNMT activity and GDM levels showed tissue-specific response. The enzymatic activity and GDM levels were lower in tumor and liver and higher in spleen and lungs under ALAN compared with controls. Our results suggest that ALAN disrupts the melatonin rhythm and potentially leading to increased BC burden by affecting DNMT activity and GDM levels. These data may also be applicable to early detection and management of BC by monitoring melatonin and GDM levels as early biomarker of ALAN circadian disruption.
Collapse
Affiliation(s)
- Sahar Agbaria
- a Department of Human Biology , University of Haifa , Haifa , Israel
| | - Abraham Haim
- b The Israeli Center for Interdisciplinary Research in Chronobiology , University of Haifa , Haifa , Israel
| | - Fuad Fares
- a Department of Human Biology , University of Haifa , Haifa , Israel.,c Department of Molecular Genetics , Carmel Medical Center , Haifa , Israel
| | - Abed E Zubidat
- b The Israeli Center for Interdisciplinary Research in Chronobiology , University of Haifa , Haifa , Israel
| |
Collapse
|
20
|
González-González A, González A, Rueda N, Alonso-González C, Menéndez-Menéndez J, Gómez-Arozamena J, Martínez-Campa C, Cos S. Melatonin Enhances the Usefulness of Ionizing Radiation: Involving the Regulation of Different Steps of the Angiogenic Process. Front Physiol 2019; 10:879. [PMID: 31354524 PMCID: PMC6637960 DOI: 10.3389/fphys.2019.00879] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/24/2019] [Indexed: 02/05/2023] Open
Abstract
Radiotherapy is a part of cancer treatment. To improve its efficacy has been combined with radiosensitizers such as antiangiogenic agents. Among the mechanisms of the antitumor action of melatonin are antiangiogenic effects. Our goal was to investigate whether melatonin may modulate the sensitivity of endothelial cells (HUVECs) to ionizing radiation. Melatonin (1 mM) enhanced the inhibition induced by radiation on different steps of the angiogenic process, cell proliferation, migration, and tubular network formation. In relation with the activity and expression of enzymes implicated in estrogen synthesis, in co-cultures HUVECs/MCF-7, radiation down-regulated aromatase mRNA expression, aromatase endothelial-specific promoter I.7, sulfatase activity and expression and 17β-HSD1 activity and expression and melatonin enhanced these effects. Radiation and melatonin induced a significant decrease in VEGF, ANG-1, and ANG-2 mRNA expression. In ANG-2 and VEGF mRNA expression melatonin potentiated the inhibitory effect induced by radiation. In addition, melatonin counteracted the stimulatory effect of radiation on FGFR3, TGFα, JAG1, IGF-1, and KDR mRNA expression and reduced ANPEP expression. In relation with extracellular matrix molecules, radiation increased MMP14 mRNA expression and melatonin counteracted the stimulatory effect of radiation on MMP14 mRNA expression and increased TIMP1 expression, an angiogenesis inhibitor. Melatonin also counteracted the stimulatory effect of radiation on CXCL6, CCL2, ERK1, ERK2, and AKT1 mRNA expression and increased the inhibitory effect of radiation on NOS3 expression. In CAM assay, melatonin enhanced the reduction of the vascular area induced by radiation. Melatonin potentiated the inhibitory effect on the activation of p-AKT and p-ERK exerted by radiation. Antiangiogenic effect of melatonin could be mediated through AKT and ERK pathways, proteins involved in vascular endothelial (VE) cell growth, cell proliferation, survival, migration, and angiogenesis. In addition, radiation increased endothelial cell permeability and melatonin counteracted it by regulating the internalization of VE-cadherin. Radiation has some side effects on angiogenesis that may reduce its effectiveness against tumor growth and melatonin is able to neutralize these negative actions of radiation. Additionally, melatonin potentiated radiation-induced antiangiogenic actions on several steps of the angiogenic process and enhanced its antitumor action. Our findings point to melatonin as a useful molecule as adjuvant to radiotherapy in cancer treatment.
Collapse
Affiliation(s)
- Alicia González-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
- *Correspondence: Alicia González,
| | - Noemí Rueda
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
| | - Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
| | - José Gómez-Arozamena
- Department of Medical Physics, School of Medicine, University of Cantabria, Santander, Spain
| | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
- Carlos Martínez-Campa,
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
| |
Collapse
|
21
|
Sonehara NM, Lacerda JZ, Jardim-Perassi BV, de Paula Jr R, Moschetta-Pinheiro MG, Souza YST, de Andrade JCJ, De Campos Zuccari DAP. Melatonin regulates tumor aggressiveness under acidosis condition in breast cancer cell lines. Oncol Lett 2018; 17:1635-1645. [PMID: 30675223 PMCID: PMC6341749 DOI: 10.3892/ol.2018.9758] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 04/26/2018] [Indexed: 12/14/2022] Open
Abstract
Breast cancer progression is composed of multiple steps that are influenced by tumor cell adaptations to survive under acidic conditions in the tumor microenvironment. Regulation of this cell survival behavior is a promising strategy to avoid cancer development. Melatonin is a natural hormone produced and secreted by the pineal gland capable of modulating different biological pathways in cancer. Although the anti-cancer effects of melatonin are currently widespread, its role in the acid tumor microenvironment remains poorly understood. The aim of the present study was to investigate the effect of low pH (6.7) on human breast cancer cell lines MCF-7 and MDA-MB-231, and the effectiveness of melatonin in acute acidosis survival mechanisms. Cell viability was measured by a MTT assay and the protein expression of glucose transporter (GLUT)-1, Ki-67 and caspase-3 was evaluated by immunocytochemical (ICC) analysis following low pH media and melatonin treatment. In both cell lines the viability was decreased after melatonin treatment (1 mM) under acidosis conditions for 24 h. ICC analysis showed a significant increase in GLUT-1 and Ki-67 expression at pH 6.7, and a decrease after treatment with melatonin for 12 and 24 h. The low pH media decreased the expression of caspase-3, which was increased after melatonin treatment for 12 and 24 h. Overall, the results of the present study revealed melatonin treatment increases apoptosis, as indicated by changes in caspase-3, and decreases proliferation, indicated by changes to Ki-67, and GLUT-1 protein expression under acute acidosis conditions in breast cancer cell lines.
Collapse
Affiliation(s)
- Nathália Martins Sonehara
- Faculty of Medicine of São José do Rio Preto (FAMERP), Department of Molecular Biology, São José do Rio Preto, SP 15090-000, Brazil.,Laboratory of Cancer Molecular Investigation (LIMC), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, SP 15090-000, Brazil
| | - Jéssica Zani Lacerda
- Laboratory of Cancer Molecular Investigation (LIMC), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, SP 15090-000, Brazil.,Sao Paulo State University 'Julio de Mesquita Filho' (UNESP), São José do Rio Preto, SP 15054-000, Brazil
| | - Bruna Victorasso Jardim-Perassi
- Faculty of Medicine of São José do Rio Preto (FAMERP), Department of Molecular Biology, São José do Rio Preto, SP 15090-000, Brazil.,Laboratory of Cancer Molecular Investigation (LIMC), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, SP 15090-000, Brazil
| | - Rubens de Paula Jr
- Faculty of Medicine of São José do Rio Preto (FAMERP), Department of Molecular Biology, São José do Rio Preto, SP 15090-000, Brazil.,Laboratory of Cancer Molecular Investigation (LIMC), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, SP 15090-000, Brazil
| | - Marina Gobbe Moschetta-Pinheiro
- Faculty of Medicine of São José do Rio Preto (FAMERP), Department of Molecular Biology, São José do Rio Preto, SP 15090-000, Brazil.,Laboratory of Cancer Molecular Investigation (LIMC), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, SP 15090-000, Brazil
| | - Ynaiá Santos Traba Souza
- Laboratory of Cancer Molecular Investigation (LIMC), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, SP 15090-000, Brazil
| | - Júlia Carolina Junqueira de Andrade
- Laboratory of Cancer Molecular Investigation (LIMC), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, SP 15090-000, Brazil
| | - Debora Aparecida Pires De Campos Zuccari
- Faculty of Medicine of São José do Rio Preto (FAMERP), Department of Molecular Biology, São José do Rio Preto, SP 15090-000, Brazil.,Laboratory of Cancer Molecular Investigation (LIMC), Department of Molecular Biology, Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto, SP 15090-000, Brazil.,Sao Paulo State University 'Julio de Mesquita Filho' (UNESP), São José do Rio Preto, SP 15054-000, Brazil
| |
Collapse
|
22
|
Santos RMD, Marani F, Chiba FY, Mattera MSDLC, Tsosura TVS, Tessarin GWL, Pereira RF, Belardi BE, Pinheiro BCES, Sumida DH. Melatonin promotes reduction in TNF levels and improves the lipid profile and insulin sensitivity in pinealectomized rats with periodontal disease. Life Sci 2018; 213:32-39. [PMID: 30321542 DOI: 10.1016/j.lfs.2018.09.056] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/25/2018] [Accepted: 09/28/2018] [Indexed: 02/04/2023]
Abstract
AIM This study aimed to investigate the effects of melatonin (ME) on insulin resistance (IR) and signaling (IS), proinflammatory cytokine levels, and lipid profiles in pinealectomyzed (PNX) rats with periodontal disease (PD). MAIN METHODS One hundred and forty-four rats (age = 40 days) were distributed into 8 groups: 1) control (CN); 2) PD only; 3) PNX only; 4) PNX and PD (PNXPD); 5) CN treated with ME (CNM); 6) PD treated with ME (PDM); 7) PNX treated with ME(PNXM); 8) PNX and PD treated with ME(PNXPDM). The PNX groups were subjected to pinealectomy at 40 and at 60 days of age. The animals were then subjected to PD induction in the mandibular first molars. After PD induction, the ME replacement therapy (MERT-5 mg/kg body weight) was performed using water for 28 days. After this period, the plasma concentration of glucose, insulin, TNF, IL-6, triglycerides, total cholesterol, HDL-cholesterol, LDL-cholesterol, and VLDL-cholesterol and the HOMA-IR index were determined. Akt serine phosphorylation status in the white adipose tissue, gastrocnemius muscle, and rat liver were also evaluated. KEY FINDINGS PD, PNX, and PNXPD groups showed an increase in IR with elevated plasma levels of insulin and TNF compared to CN group. PNX and PNXPD groups presented alteration in lipid profile compared to CN group. MERT improved all of the analyzed parameters. No difference was observed in the IS among different groups. SIGNIFICANCE The results suggest that MERT efficiently prevents IR, improves lipid profile, and increases plasma levels of insulin and TNF in PD and PNX rats.
Collapse
Affiliation(s)
- Rodrigo Martins Dos Santos
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas-SBFis, Brazil; Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Fernando Marani
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas-SBFis, Brazil; Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Fernando Yamamoto Chiba
- Preventive and Social Dentistry Post-Graduation Program School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Maria Sara de Lima Coutinho Mattera
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas-SBFis, Brazil; Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Thais Verônica Saori Tsosura
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas-SBFis, Brazil; Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Gestter Willian Lattari Tessarin
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil; Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Renato Felipe Pereira
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas-SBFis, Brazil; Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Bianca Elvira Belardi
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Beatriz Costa E Silva Pinheiro
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil
| | - Doris Hissako Sumida
- Programa de Pós-graduação Multicêntrico em Ciências Fisiológicas-SBFis, Brazil; Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), Araçatuba, SP, Brazil.
| |
Collapse
|
23
|
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
|
24
|
González-González A, Mediavilla MD, Sánchez-Barceló EJ. Melatonin: A Molecule for Reducing Breast Cancer Risk. Molecules 2018; 23:E336. [PMID: 29415446 PMCID: PMC6017232 DOI: 10.3390/molecules23020336] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 01/31/2018] [Accepted: 01/31/2018] [Indexed: 01/10/2023] Open
Abstract
The objective of this article is to review the basis supporting the usefulness of melatonin as an adjuvant therapy for breast cancer (BC) prevention in several groups of individuals at high risk for this disease. Melatonin, as a result of its antiestrogenic and antioxidant properties, as well as its ability to improve the efficacy and reduce the side effects of conventional antiestrogens, could safely be associated with the antiestrogenic drugs presently in use. In individuals at risk of BC due to night shift work, the light-induced inhibition of melatonin secretion, with the consequent loss of its antiestrogenic effects, would be countered by administering this neurohormone. BC risk from exposure to metalloestrogens, such as cadmium, could be treated with melatonin supplements to individuals at risk of BC due to exposure to this xenoestrogen. The BC risk related to obesity may be reduced by melatonin which decrease body fat mass, inhibits the enhanced aromatase expression in obese women, increases adiponectin secretion, counteracts the oncogenic effects of elevated concentrations of leptin; and decreases blood glucose levels and insulin resistance. Despite compelling experimental evidence of melatonin's oncostatic actions being susceptible to lowering BC risk, there is still a paucity of clinical trials focused on this subject.
Collapse
Affiliation(s)
- Alicia González-González
- Department of Physiology and Pharmacology, School of Medicina, University of Cantabria, 39011 Santander, Spain.
| | - María Dolores Mediavilla
- Department of Physiology and Pharmacology, School of Medicina, University of Cantabria, 39011 Santander, Spain.
| | - Emilio J Sánchez-Barceló
- Department of Physiology and Pharmacology, School of Medicina, University of Cantabria, 39011 Santander, Spain.
| |
Collapse
|
25
|
Johns LE, Jones ME, Schoemaker MJ, McFadden E, Ashworth A, Swerdlow AJ. Domestic light at night and breast cancer risk: a prospective analysis of 105 000 UK women in the Generations Study. Br J Cancer 2018; 118:600-606. [PMID: 29360812 PMCID: PMC5830585 DOI: 10.1038/bjc.2017.359] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/06/2017] [Accepted: 09/14/2017] [Indexed: 01/09/2023] Open
Abstract
Background: Circadian disruption caused by exposure to light at night (LAN) has been proposed as a risk factor for breast cancer and a reason for secular increases in incidence. Studies to date have largely been ecological or case-control in design and findings have been mixed. Methods: We investigated the relationship between LAN and breast cancer risk in the UK Generations Study. Bedroom light levels and sleeping patterns at age 20 and at study recruitment were obtained by questionnaire. Analyses were conducted on 105 866 participants with no prior history of breast cancer. During an average of 6.1 years of follow-up, 1775 cases of breast cancer were diagnosed. Cox proportional hazard models were used to calculate hazard ratios (HRs), adjusting for potential confounding factors. Results: There was no association between LAN level and breast cancer risk overall (highest compared with lowest LAN level at recruitment: HR=1.01, 95% confidence interval (CI): 0.88–1.15), or for invasive (HR=0.98, 95% CI: 0.85–1.13) or in situ (HR=0.96, 95% CI: 0.83–1.11) breast cancer, or oestrogen-receptor (ER) positive (HR=0.98, 95% CI: 0.84–1.14); or negative (HR=1.16, 95% CI: 0.82–1.65) tumours separately. The findings did not differ by menopausal status. Adjusting for sleep duration, sleeping at unusual times (non-peak sleep) and history of night work did not affect the results. Night waking with exposure to light, occurring around age 20, was associated with a reduced risk of premenopausal breast cancer (HR for breast cancer overall=0.74, 95% CI: 0.55–0.99; HR for ER-positive breast cancer=0.69, 95% CI: 0.49–0.97). Conclusions: In this prospective cohort analysis of LAN, there was no evidence that LAN exposure increased the risk of subsequent breast cancer, although the suggestion of a lower breast cancer risk in pre-menopausal women with a history of night waking in their twenties may warrant further investigation.
Collapse
Affiliation(s)
- Louise E Johns
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Michael E Jones
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Minouk J Schoemaker
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Emily McFadden
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SM2 5NG, UK
| | - Alan Ashworth
- Division of Breast Cancer Research, The Institute of Cancer Research, London SW3 6JB, UK.,Breast Cancer Now Research Centre at The Institute of Cancer Research, London SW3 6JB, UK.,Division of Molecular Pathology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SM2 5NG, UK.,Division of Breast Cancer Research, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
26
|
Gray JM, Rasanayagam S, Engel C, Rizzo J. State of the evidence 2017: an update on the connection between breast cancer and the environment. Environ Health 2017; 16:94. [PMID: 28865460 PMCID: PMC5581466 DOI: 10.1186/s12940-017-0287-4] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 07/17/2017] [Indexed: 05/23/2023]
Abstract
BACKGROUND In this review, we examine the continually expanding and increasingly compelling data linking radiation and various chemicals in our environment to the current high incidence of breast cancer. Singly and in combination, these toxicants may have contributed significantly to the increasing rates of breast cancer observed over the past several decades. Exposures early in development from gestation through adolescence and early adulthood are particularly of concern as they re-shape the program of genetic, epigenetic and physiological processes in the developing mammary system, leading to an increased risk for developing breast cancer. In the 8 years since we last published a comprehensive review of the relevant literature, hundreds of new papers have appeared supporting this link, and in this update, the evidence on this topic is more extensive and of better quality than that previously available. CONCLUSION Increasing evidence from epidemiological studies, as well as a better understanding of mechanisms linking toxicants with development of breast cancer, all reinforce the conclusion that exposures to these substances - many of which are found in common, everyday products and byproducts - may lead to increased risk of developing breast cancer. Moving forward, attention to methodological limitations, especially in relevant epidemiological and animal models, will need to be addressed to allow clearer and more direct connections to be evaluated.
Collapse
Affiliation(s)
- Janet M. Gray
- Department of Psychology and Program in Science, Technology, and Society, Vassar College, 124 Raymond Avenue, Poughkeepsie, NY 12604-0246 USA
| | - Sharima Rasanayagam
- Breast Cancer Prevention Partners, 1388 Sutter St., Suite 400, San Francisco, CA 94109-5400 USA
| | - Connie Engel
- Breast Cancer Prevention Partners, 1388 Sutter St., Suite 400, San Francisco, CA 94109-5400 USA
| | - Jeanne Rizzo
- Breast Cancer Prevention Partners, 1388 Sutter St., Suite 400, San Francisco, CA 94109-5400 USA
| |
Collapse
|
27
|
González A, González-González A, Alonso-González C, Menéndez-Menéndez J, Martínez-Campa C, Cos S. Melatonin inhibits angiogenesis in SH-SY5Y human neuroblastoma cells by downregulation of VEGF. Oncol Rep 2017; 37:2433-2440. [PMID: 28259965 DOI: 10.3892/or.2017.5446] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/10/2016] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) produced from tumor cells plays a crucial role in the pathogenesis and neovascularization of neuroblastoma. Inhibition of VEGF secretion by tumor cells, as well as VEGF-regulated signaling in endothelial cells, are important to reduce the angiogenesis and growth of neuroblastoma. Since melatonin has anti-angiogenic effects in tumor cell lines, the aim of the present study was to study melatonin modulation of the pro-angiogenic effects of VEGF in neuroblastoma cells (SH-SY5Y). We used co-cultures of SH-SY5Y and endothelial cells. VEGF expression and protein levels were analyzed by quantitative RT-PCR and ELISA, respectively. Endothelial cell migration was assessed by wound-healing assay and endothelial angiogenesis by a tube formation assay. Melatonin inhibited the pro-angiogenic effects of SH-SY5Y cells. The conditioned medium collected from the neuroblastoma cells was angiogenically active and stimulated proliferation, migration and tube formation in endothelial cells. This effect was significantly counteracted by the addition of either anti-VEGF or melatonin. Melatonin inhibited VEGF expression and secretion in SH-SY5Y cells, decreasing the levels of VEGF available for endothelial cells. Melatonin has anti-angiogenic effects at different steps of the angiogenic process in SH-SY5Y neuroblastoma cells, through the downregulation of VEGF.
Collapse
Affiliation(s)
- Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute (IDIVAL), ES-39011 Santander, Spain
| | - Alicia González-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute (IDIVAL), ES-39011 Santander, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute (IDIVAL), ES-39011 Santander, Spain
| | - Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute (IDIVAL), ES-39011 Santander, Spain
| | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute (IDIVAL), ES-39011 Santander, Spain
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute (IDIVAL), ES-39011 Santander, Spain
| |
Collapse
|
28
|
Gupta P, Pushkala K. Increased Incidence of Breast Cancer Due to Long Exposure of Light. ACTA ACUST UNITED AC 2016. [DOI: 10.6000/1927-7229.2016.05.04.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
29
|
Melatonin promotes ATO-induced apoptosis in MCF-7 cells: Proposing novel therapeutic potential for breast cancer. Biomed Pharmacother 2016; 83:456-465. [DOI: 10.1016/j.biopha.2016.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/29/2016] [Accepted: 07/03/2016] [Indexed: 02/06/2023] Open
|
30
|
Ball LJ, Palesh O, Kriegsfeld LJ. The Pathophysiologic Role of Disrupted Circadian and Neuroendocrine Rhythms in Breast Carcinogenesis. Endocr Rev 2016; 37:450-466. [PMID: 27712099 PMCID: PMC5045494 DOI: 10.1210/er.2015-1133] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most physiological processes in the brain and body exhibit daily (circadian) rhythms coordinated by an endogenous master clock located in the suprachiasmatic nucleus of the hypothalamus that are essential for normal health and functioning. Exposure to sunlight during the day and darkness at night optimally entrains biological rhythms to promote homeostasis and human health. Unfortunately, a major consequence of the modern lifestyle is increased exposure to sun-free environments during the day and artificial lighting at night. Additionally, behavioral disruptions to circadian rhythms (ie, repeated transmeridian flights, night or rotating shift work, or sleep disturbances) have a profound influence on health and have been linked to a number of pathological conditions, including endocrine-dependent cancers. Specifically, night shift work has been identified as a significant risk factor for breast cancer in industrialized countries. Several mechanisms have been proposed by which shift work-induced circadian disruptions promote cancer. In this review, we examine the importance of the brain-body link through which circadian disruptions contribute to endocrine-dependent diseases, including breast carcinogenesis, by negatively impacting neuroendocrine and neuroimmune cells, and we consider preventive measures directed at maximizing circadian health.
Collapse
Affiliation(s)
- Lonnele J Ball
- Department of Psychology (L.J.B., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, California 94720; and Department of Psychiatry and Behavioral Sciences (O.P.), Stanford University School of Medicine, Stanford, California 94305
| | - Oxana Palesh
- Department of Psychology (L.J.B., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, California 94720; and Department of Psychiatry and Behavioral Sciences (O.P.), Stanford University School of Medicine, Stanford, California 94305
| | - Lance J Kriegsfeld
- Department of Psychology (L.J.B., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, California 94720; and Department of Psychiatry and Behavioral Sciences (O.P.), Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
31
|
Lin YW, Lee LM, Lee WJ, Chu CY, Tan P, Yang YC, Chen WY, Yang SF, Hsiao M, Chien MH. Melatonin inhibits MMP-9 transactivation and renal cell carcinoma metastasis by suppressing Akt-MAPKs pathway and NF-κB DNA-binding activity. J Pineal Res 2016; 60:277-90. [PMID: 26732239 DOI: 10.1111/jpi.12308] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/04/2016] [Indexed: 12/13/2022]
Abstract
Renal cell carcinoma (RCC) is the most lethal of all urological malignancies because of its potent metastasis potential. Melatonin exerts multiple tumor-suppressing activities through antiproliferative, proapoptotic, and anti-angiogenic actions and has been tested in clinical trials. However, the antimetastastic effect of melatonin and its underlying mechanism in RCC are unclear. In this study, we demonstrated that melatonin at the pharmacologic concentration (0.5-2 mm) considerably reduced the migration and invasion of RCC cells (Caki-1 and Achn). Furthermore, we found that melatonin suppressed metastasis of Caki-1 cells in spontaneous and experimental metastasis animal models. Mechanistic investigations revealed that melatonin transcriptionally inhibited MMP-9 by reducing p65- and p52-DNA-binding activities. Moreover, the Akt-mediated JNK1/2 and ERK1/2 signaling pathways were involved in melatonin-regulated MMP-9 transactivation and cell motility. Clinical samples revealed an inverse correlation between melatonin receptor 1A (MTNR1A) and MMP-9 expression in normal kidney and RCC tissues. In addition, a higher survival rate was found in MTNR1A(high) /MMP-9(low) patients than in MTNR1A(low) /MMP-9(high) patients. Overall, our results provide new insights into the role of melatonin-induced molecular regulation in suppressing RCC metastasis and suggest that melatonin has potential therapeutic applications for metastastic RCC.
Collapse
Affiliation(s)
- Yung-Wei Lin
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Liang-Ming Lee
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ying Chu
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Peng Tan
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chieh Yang
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Yu Chen
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
32
|
Melatonin, an inhibitory agent in breast cancer. Breast Cancer 2016; 24:42-51. [PMID: 27017208 DOI: 10.1007/s12282-016-0690-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/15/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND The heterogeneous nature of breast cancer makes it one of the most challenging cancers to treat. Due to the stimulatory effect of estrogen in mammary cancer progression, anti-estrogenic agents like melatonin have found their way into breast cancer treatment. Further studies confirmed a reverse correlation between nocturnal melatonin levels and the development of mammary cancer. In this study we reviewed the molecular inhibitory effects of melatonin in breast cancer therapy. METHODS To open access the articles, Google scholar and science direct were used as a motor search. We used from valid external and internal databases. To reach the search formula, we determined mean key words like breast cancer, melatonin, cell proliferation and death. To retrieval the related articles, we continuously search the articles from 1984 to 2015. The relevance and the quality of the 480 articles were screened; at least we selected 80 eligible articles about melatonin molecular mechanism in breast cancer. RESULT The results showed that melatonin not only inhibits breast cancer cell growth, but also is capable of inhibiting angiogenesis, cancer cell invasion, and telomerase activity. Interestingly this hormone is able to induce apoptosis through the suppression or induction of a wide range of signaling pathways. Moreover, it seems that the concomitant administration of melatonin with other conventional chemotherapy agents had beneficial effects for patients with breast cancer, by alleviating unfavorable effects of those agents and enhancing their efficacy. CONCLUSION The broad inhibitory effects of melatonin in breast cancer make it a promising agent and may add it to the list of potential drugs in treatment of this cancer.
Collapse
|
33
|
França EL, Honorio-França AC, Fernandes RTDS, Marins CMF, Pereira CCDS, Varotti FDP. The Effect of Melatonin Adsorbed to Polyethylene Glycol Microspheres on the Survival of MCF-7 Cells. Neuroimmunomodulation 2016; 23:27-32. [PMID: 26445481 DOI: 10.1159/000439277] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/06/2015] [Indexed: 11/19/2022] Open
Abstract
Although melatonin exhibits oncostatic properties such as antiproliferative effects, the oral bioavailability of this hormone is less than 20%. Modified drug release systems have been used to improve the pharmacological efficiency of drugs. These systems can change the pharmacokinetics and biodistribution of the associated drugs. Thus, this study investigated the effect of melatonin adsorbed to polyethylene glycol (PEG) microspheres on MCF-7 human breast cancer cells. The MCF-7 cells were obtained from the American Type Culture Collection. MCF-7 cells were preincubated for 24 h with or without melatonin (100 ng/ml), PEG microspheres or melatonin adsorbed to PEG microspheres (100 ng/ml). Viability, intracellular calcium release and apoptosis in MCF-7 cells were determined by flow cytometry. MCF-7 cells incubated with melatonin adsorbed to PEG microspheres showed a lower viability rate (40.0 ± 8.3 with melatonin adsorbed to PEG microspheres compared to 54.1 ± 7.3 with melatonin; 81.8 ± 12.5 with PEG microsphere and 92.7 ± 4.1 with medium), increased spontaneous intracellular Ca2+ release (27.0 ± 8.6 with melatonin adsorbed to PEG microspheres compared to 21.5 ± 13.4 with melatonin; 10.1 ± 5.4 with PEG microsphere and 9.1 ± 5.6 with medium) and increased apoptosis index (51.2 ± 2.7 with melatonin adsorbed to PEG microspheres compared to 36.0 ± 2.1 with melatonin; 4.9 ± 0.5 with PEG microsphere and 3.1 ± 0.6 with medium). The results indicate that melatonin adsorbed to PEG microspheres exerts antitumor effects on human MCF-7 breast cancer cells. However, clinical tests must be performed to confirm the use of melatonin adsorbed to PEG microspheres as an alternative therapy against cancer.
Collapse
Affiliation(s)
- Eduardo Luzía França
- Institute of Biological and Health Science, Federal University of Mato Grosso, Barra do Garx00E7;as, Brazil
| | | | | | | | | | | |
Collapse
|
34
|
Alonso-González C, González A, Martínez-Campa C, Menéndez-Menéndez J, Gómez-Arozamena J, García-Vidal A, Cos S. Melatonin enhancement of the radiosensitivity of human breast cancer cells is associated with the modulation of proteins involved in estrogen biosynthesis. Cancer Lett 2016; 370:145-52. [PMID: 26497762 DOI: 10.1016/j.canlet.2015.10.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/14/2015] [Accepted: 10/15/2015] [Indexed: 02/07/2023]
Abstract
Enhancing the radiosensitivity of cancer cells is one of the most important tasks in clinical radiobiology. Endocrine therapy and radiotherapy are two cancer treatment modalities which are often given together in patients with locally-advanced breast cancer and positive hormone-receptor status. Oncostatic actions of melatonin are relevant on estrogen-dependent mammary tumors. In the present study, we wanted to evaluate the effects of the combination of ionizing radiation and melatonin on proteins involved in estrogen biosynthesis in breast cancer cells. We demonstrated a role of melatonin in mediating the sensitization of human breast cancer cells to the ionizing radiation by decreasing around 50% the activity and expression of proteins involved in the synthesis of estrogens in these cells. Thus, melatonin pretreatment before radiation reduces the amount of active estrogens at cancer cell level. Melatonin 1 nM induced a 2-fold change in p53 expression as compared to radiation alone. The regulatory action of melatonin on p53 could be a link between melatonin and its modulatory action on the sensitivity of breast cancer cells to ionizing radiation. These findings may have implications for designing clinical trials using melatonin and radiotherapy.
Collapse
Affiliation(s)
- Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), Spain
| | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), Spain
| | - Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), Spain
| | - José Gómez-Arozamena
- Department of Medical Physics, School of Medicine, University of Cantabria, 39011 Santander, Spain
| | - Angela García-Vidal
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), Spain
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), Spain.
| |
Collapse
|
35
|
GONZÁLEZ ALICIA, MARTÍNEZ-CAMPA CARLOS, ALONSO-GONZÁLEZ CAROLINA, COS SAMUEL. Melatonin affects the dynamic steady-state equilibrium of estrogen sulfates in human umbilical vein endothelial cells by regulating the balance between estrogen sulfatase and sulfotransferase. Int J Mol Med 2015; 36:1671-6. [DOI: 10.3892/ijmm.2015.2360] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/25/2015] [Indexed: 11/05/2022] Open
|
36
|
Hill SM, Belancio VP, Dauchy RT, Xiang S, Brimer S, Mao L, Hauch A, Lundberg PW, Summers W, Yuan L, Frasch T, Blask DE. Melatonin: an inhibitor of breast cancer. Endocr Relat Cancer 2015; 22:R183-204. [PMID: 25876649 PMCID: PMC4457700 DOI: 10.1530/erc-15-0030] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2015] [Indexed: 12/19/2022]
Abstract
The present review discusses recent work on melatonin-mediated circadian regulation, the metabolic and molecular signaling mechanisms that are involved in human breast cancer growth, and the associated consequences of circadian disruption by exposure to light at night (LEN). The anti-cancer actions of the circadian melatonin signal in human breast cancer cell lines and xenografts heavily involve MT1 receptor-mediated mechanisms. In estrogen receptor alpha (ERα)-positive human breast cancer, melatonin suppresses ERα mRNA expression and ERα transcriptional activity via the MT1 receptor. Melatonin also regulates the transactivation of other members of the nuclear receptor superfamily, estrogen-metabolizing enzymes, and the expression of core clock and clock-related genes. Furthermore, melatonin also suppresses tumor aerobic metabolism (the Warburg effect) and, subsequently, cell-signaling pathways critical to cell proliferation, cell survival, metastasis, and drug resistance. Melatonin demonstrates both cytostatic and cytotoxic activity in breast cancer cells that appears to be cell type-specific. Melatonin also possesses anti-invasive/anti-metastatic actions that involve multiple pathways, including inhibition of p38 MAPK and repression of epithelial-mesenchymal transition (EMT). Studies have demonstrated that melatonin promotes genomic stability by inhibiting the expression of LINE-1 retrotransposons. Finally, research in animal and human models has indicated that LEN-induced disruption of the circadian nocturnal melatonin signal promotes the growth, metabolism, and signaling of human breast cancer and drives breast tumors to endocrine and chemotherapeutic resistance. These data provide the strongest understanding and support of the mechanisms that underpin the epidemiologic demonstration of elevated breast cancer risk in night-shift workers and other individuals who are increasingly exposed to LEN.
Collapse
Affiliation(s)
- Steven M Hill
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Victoria P Belancio
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Robert T Dauchy
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Shulin Xiang
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Samantha Brimer
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Lulu Mao
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Adam Hauch
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Peter W Lundberg
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Whitney Summers
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Lin Yuan
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - Tripp Frasch
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | - David E Blask
- Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA Department of Structural and Cellular BiologyTulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, Louisiana 70112, USADepartment of SurgeryTulane Cancer Center and Louisiana Cancer Research ConsortiumCircadian Cancer Biology GroupTulane Center for Circadian BiologyTulane University School of Medicine, New Orleans, Louisiana 70112, USA
| |
Collapse
|
37
|
Squecco R, Tani A, Zecchi-Orlandini S, Formigli L, Francini F. Melatonin affects voltage-dependent calcium and potassium currents in MCF-7 cell line cultured either in growth or differentiation medium. Eur J Pharmacol 2015; 758:40-52. [PMID: 25843408 DOI: 10.1016/j.ejphar.2015.03.068] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/24/2015] [Accepted: 03/25/2015] [Indexed: 11/29/2022]
Abstract
Big efforts have been dedicated up to now to identify novel targets for cancer treatment. The peculiar biophysical profile and the atypical ionic channels activity shown by diverse types of human cancers suggest that ion channels may be possible targets in cancer therapy. Earlier studies have shown that melatonin exerts an oncostatic action on different tumors. In particular, it was shown that melatonin was able to inhibit growth/viability and proliferation, to reduce the invasiveness and metastatic properties of human estrogen-sensitive breast adenocarcinoma MCF-7 cell line cultured in growth medium, with substantial impairments of epidermal growth factor (EGF) and Notch-1-mediated signaling. The purpose of this work was to evaluate on MCF-7 cells the possible effects of melatonin on the biophysical features known to have a role in proliferation and differentiation, by using the patch-clamp technique. Our results show that in cells cultured in growth as well as in differentiation medium melatonin caused a hyperpolarization of resting membrane potential paralleled by significant changes of the inward Ca(2+) currents (T- and L-type), outward delayed rectifier K(+) currents and cell capacitance. All these effects are involved in MCF-7 growth and differentiation. These findings strongly suggest that melatonin, acting as a modulator of different voltage-dependent ion channels, might be considered a new promising tool for specifically disrupting cell viability and differentiation pathways in tumour cells with possible beneficial effects on cancer therapy.
Collapse
Affiliation(s)
- Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy
| | - Lucia Formigli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy
| | - Fabio Francini
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy.
| |
Collapse
|
38
|
Alonso-González C, González A, Martínez-Campa C, Gómez-Arozamena J, Cos S. Melatonin sensitizes human breast cancer cells to ionizing radiation by downregulating proteins involved in double-strand DNA break repair. J Pineal Res 2015; 58:189-97. [PMID: 25623566 DOI: 10.1111/jpi.12205] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 01/06/2015] [Indexed: 11/28/2022]
Abstract
Radiation and adjuvant endocrine therapy are nowadays considered a standard treatment option after surgery in breast cancer. Melatonin exerts oncostatic actions on human breast cancer cells. In the current study, we investigated the effects of a combination of radiotherapy and melatonin on human breast cancer cells. Melatonin (1 mm, 10 μm and 1 nm) significantly inhibited the proliferation of MCF-7 cells. Radiation alone inhibited the MCF-7 cell proliferation in a dose-dependent manner. Pretreatment of breast cancer cells with melatonin 1 wk before radiation led to a significantly greater decrease of MCF-7 cell proliferation compared with radiation alone. Melatonin pretreatment before radiation also decreased G2 -M phase arrest compared with irradiation alone, with a higher percentage of cells in the G0 -G1 phase and a lower percentage of cells in S phase. Radiation alone diminished RAD51 and DNA-protein kinase (PKcs) mRNA expression, two main proteins involved in double-strand DNA break repair. Treatment with melatonin for 7 days before radiation led to a significantly greater decrease in RAD51 and DNA-PKcs mRNA expression compared with radiation alone. Our findings suggest that melatonin pretreatment before radiation sensitizes breast cancer cells to the ionizing effects of radiation by decreasing cell proliferation, inducing cell cycle arrest and downregulating proteins involved in double-strand DNA break repair. These findings may have implications for designing clinical trials using melatonin and radiotherapy.
Collapse
Affiliation(s)
- Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Valdecilla (IDIVAL), Santander, Spain
| | | | | | | | | |
Collapse
|
39
|
Arnao MB, Hernández-Ruiz J. Phytomelatonin: Searching for Plants with High Levels for Use as a Natural Nutraceutical. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2015. [DOI: 10.1016/b978-0-444-63462-7.00011-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
40
|
Rabstein S, Harth V, Justenhoven C, Pesch B, Plöttner S, Heinze E, Lotz A, Baisch C, Schiffermann M, Brauch H, Hamann U, Ko Y, Brüning T. Polymorphisms in circadian genes, night work and breast cancer: results from the GENICA study. Chronobiol Int 2014; 31:1115-22. [PMID: 25229211 DOI: 10.3109/07420528.2014.957301] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES The role of genetic variants and environmental factors in breast cancer etiology has been intensively studied in the last decades. Gene-environment interactions are now increasingly being investigated to gain more insights into the development of breast cancer, specific subtypes, and therapeutics. Recently, night shift work that involves circadian disruption has gained rising interest as a potential non-genetic breast cancer risk factor. Here, we analyzed genetic polymorphisms in genes of cellular clocks, melatonin biosynthesis and signaling and their association with breast cancer as well as gene-gene and gene-night work interactions in a German case-control study on breast cancer. METHODS GENICA is a population-based case-control study on breast cancer conducted in the Greater Region of Bonn. Associations between seven polymorphisms in circadian genes (CLOCK, NPAS2, ARTNL, PER2 and CRY2), genes of melatonin biosynthesis and signaling (AANAT and MTNR1B) and breast cancer were analyzed with conditional logistic regression models, adjusted for potential confounders for 1022 cases and 1014 controls. Detailed shift-work information was documented for 857 breast cancer cases and 892 controls. Gene-gene and gene-shiftwork interactions were analyzed using model-based multifactor dimensionality reduction (mbMDR). RESULTS For combined heterozygotes and rare homozygotes a slightly elevated breast cancer risk was found for rs8150 in gene AANAT (OR 1.17; 95% CI 1.01-1.36), and a reduced risk for rs3816358 in gene ARNTL (OR 0.82; 95% CI 0.69-0.97) in the complete study population. In the subgroup of shift workers, rare homozygotes for rs10462028 in the CLOCK gene had an elevated risk of breast cancer (OR for AA vs. GG: 3.53; 95% CI 1.09-11.42). Shift work and CLOCK gene interactions were observed in the two-way interaction analysis. In addition, gene-shiftwork interactions were detected for MTNR1B with NPAS2 and ARNTL. CONCLUSIONS In conclusion, the results of our population-based case-control study support a putative role of the CLOCK gene in the development of breast cancer in shift workers. In addition, higher order interaction analyses suggest a potential relevance of MTNR1B with the key transcriptional factor NPAS2 with ARNTL. Hence, in the context of circadian disruption, multivariable models should be preferred that consider a wide range of polymorphisms, e.g. that may influence chronotype or light sensitivity. The investigation of these interactions in larger studies is needed.
Collapse
Affiliation(s)
- Sylvia Rabstein
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA) , Bochum , Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chottanapund S, Van Duursen MBM, Navasumrit P, Hunsonti P, Timtavorn S, Ruchirawat M, Van den Berg M. Anti-aromatase effect of resveratrol and melatonin on hormonal positive breast cancer cells co-cultured with breast adipose fibroblasts. Toxicol In Vitro 2014; 28:1215-21. [PMID: 24929094 DOI: 10.1016/j.tiv.2014.05.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 05/27/2014] [Accepted: 05/29/2014] [Indexed: 12/29/2022]
Abstract
Targeting the estrogen pathway has been proven effective in the treatment for estrogen receptor positive breast cancer. There are currently two common groups of anti-estrogenic compounds used in the clinic; Selective Estrogen Receptor Modulators (SERMs, e.g. tamoxifen) and Selective Estrogen Enzyme Modulators (SEEMs e.g. letrozole). Among various naturally occurring, biologically active compounds, resveratrol and melatonin have been suggested to act as aromatase inhibitors, which make them potential candidates in hormonal treatment of breast cancer. Here we used a co-culture model in which we previously demonstrated that primary human breast adipose fibroblasts (BAFs) can convert testosterone to estradiol, which subsequently results in estrogen receptor-mediated breast cancer T47D cell proliferation. In the presence of testosterone in this model, we examined the effect of letrozole, resveratrol and melatonin on cell proliferation, estradiol (E2) production and gene expression of CYP19A1, pS2 and Ki-67. Both melatonin and resveratrol were found to be aromatase inhibitors in this co-culture system, albeit at different concentrations. Our co-culture model did not provide any indications that melatonin is also a selective estrogen receptor modulator. In the T47D-BAF co-culture, a melatonin concentration of 20 nM and resveratrol concentration of 20 μM have an aromatase inhibitory effect as potent as 20 nM letrozole, which is a clinically used anti-aromatase drug in breast cancer treatment. The SEEM mechanism of action of especially melatonin clearly offers potential advantages for breast cancer treatment.
Collapse
Affiliation(s)
- Suthat Chottanapund
- Division of Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand; Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand; Center of Excellence on Environmental Health, Toxicology and Management of Chemicals, Bangkok, Thailand; Bamrasnaradura Infectious Diseases Institute, Ministry of Public Health, Thailand.
| | - M B M Van Duursen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Panida Navasumrit
- Division of Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand; Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand; Center of Excellence on Environmental Health, Toxicology and Management of Chemicals, Bangkok, Thailand
| | - Potchanee Hunsonti
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Supatchaya Timtavorn
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Mathuros Ruchirawat
- Division of Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand; Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand; Center of Excellence on Environmental Health, Toxicology and Management of Chemicals, Bangkok, Thailand
| | - Martin Van den Berg
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
42
|
Cos S, Alvarez-García V, González A, Alonso-González C, Martínez-Campa C. Melatonin modulation of crosstalk among malignant epithelial, endothelial and adipose cells in breast cancer (Review). Oncol Lett 2014; 8:487-492. [PMID: 25009641 PMCID: PMC4081418 DOI: 10.3892/ol.2014.2203] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 05/13/2014] [Indexed: 11/29/2022] Open
Abstract
Melatonin, the main secretory product of the pineal gland, is an oncostatic agent that reduces the growth and development of various types of tumors, particularly mammary tumors whose growth is dependent on estrogens. Previous in vivo and in vitro studies point to the hypothesis that melatonin interplays with estrogen signaling pathways at three different levels: i) an indirect mechanism, by interfering with the hypothalamic-pituitary-reproductive axis in such way that the level of plasma estrogens synthesized by the gonadal glands are downregulated; ii) a direct mechanism of the pineal gland at the cell cancer level, disrupting the activation of estradiol receptors, therefore behaving as a selective estrogen receptor modulator; and iii) by regulating the enzymes involved in the biosynthesis of estrogens in other tissues, thus behaving as a selective estrogen enzyme modulator. The intratumoral metabolism and synthesis of estrogens, as a result of the interactions of various enzymes, is more important than blood uptake to maintain mammary gland estrogen levels in menopausal females. Additionally, estrogens are considered to play an important role in the pathogenesis and development of hormone-dependent breast carcinoma. Paracrine interactions among malignant epithelial cells and proximal adipose and endothelial cells, through cytokines and growth factors produced by breast tumor cells, modulate estrogen production at the mammary tumor level and, as a consequence, the genesis and development of mammary tumors. The aim of the present review is to summarize the recent findings describing the mechanisms by which melatonin is able to modulate the crosstalk among malignant epithelial, endothelial and adipose cells in breast cancer.
Collapse
Affiliation(s)
- Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute, Santander 39011, Spain
| | - Virginia Alvarez-García
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute, Santander 39011, Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute, Santander 39011, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute, Santander 39011, Spain
| | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Valdecilla Research Institute, Santander 39011, Spain
| |
Collapse
|
43
|
Chen WY, Giobbie-Hurder A, Gantman K, Savoie J, Scheib R, Parker LM, Schernhammer ES. A randomized, placebo-controlled trial of melatonin on breast cancer survivors: impact on sleep, mood, and hot flashes. Breast Cancer Res Treat 2014; 145:381-8. [PMID: 24718775 DOI: 10.1007/s10549-014-2944-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 11/29/2022]
Abstract
The purpose is to examine the effects of melatonin supplementation on sleep, mood, and hot flashes in postmenopausal breast cancer survivors. In a randomized, double-blind, placebo-controlled study, 95 postmenopausal women with a prior history of stage 0-III breast cancer, who had completed active cancer treatment (including hormonal therapy) were randomly assigned 1:1 to either 3 mg oral melatonin (n = 48) or placebo daily (n = 47) for 4 months. Sleep, mood, and hot flashes were assessed at baseline and 4 months via self-administered questionnaire using the Pittsburgh Sleep Quality Index (PSQI), Center for Epidemiologic Studies-Depression (CES-D), and the North Central Cancer Treatment Group (NCCTG) hot flash diary, respectively. Eighty-six women (91 %) completed the study and provided pre- and post-questionnaires. At baseline, 52 % of participants reported poor sleep in the month prior to enrollment. Compared to subjects on placebo, subjects randomized to melatonin experienced significantly greater improvements in subjective sleep quality as measured by the PSQI, including domains on sleep quality, daytime dysfunction and total score. For example, the mean change in PSQI score was -0.1 in the placebo group compared to -1.9 in the melatonin group (p < 0.001). There were no significant differences in measures of depression or hot flashes. Sleep disturbances are common among breast cancer survivors, even after completion of active cancer treatment. This is the first randomized placebo-controlled study among breast cancer survivors to demonstrate that melatonin was associated with an improvement in subjective sleep quality, without any significant adverse effects.
Collapse
Affiliation(s)
- Wendy Y Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA,
| | | | | | | | | | | | | |
Collapse
|
44
|
García JJ, López-Pingarrón L, Almeida-Souza P, Tres A, Escudero P, García-Gil FA, Tan DX, Reiter RJ, Ramírez JM, Bernal-Pérez M. Protective effects of melatonin in reducing oxidative stress and in preserving the fluidity of biological membranes: a review. J Pineal Res 2014; 56:225-37. [PMID: 24571249 DOI: 10.1111/jpi.12128] [Citation(s) in RCA: 336] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 02/20/2014] [Indexed: 12/14/2022]
Abstract
Free radicals generated within subcellular compartments damage macromolecules which lead to severe structural changes and functional alterations of cellular organelles. A manifestation of free radical injury to biological membranes is the process of lipid peroxidation, an autooxidative chain reaction in which polyunsaturated fatty acids in the membrane are the substrate. There is considerable evidence that damage to polyunsaturated fatty acids tends to reduce membrane fluidity. However, adequate levels of fluidity are essential for the proper functioning of biological membranes. Thus, there is considerable interest in antioxidant molecules which are able to stabilize membranes because of their protective effects against lipid peroxidation. Melatonin is an indoleamine that modulates a wide variety of endocrine, neural and immune functions. Over the last two decades, intensive research has proven this molecule, as well as its metabolites, to possess substantial antioxidant activity. In addition to their ability to scavenge several reactive oxygen and nitrogen species, melatonin increases the activity of the glutathione redox enzymes, that is, glutathione peroxidase and reductase, as well as other antioxidant enzymes. These beneficial effects of melatonin are more significant because of its small molecular size and its amphipathic behaviour, which facilitates ease of melatonin penetration into every subcellular compartment. In the present work, we review the current information related to the beneficial effects of melatonin in maintaining the fluidity of biological membranes against free radical attack, and further, we discuss its implications for ageing and disease.
Collapse
Affiliation(s)
- Joaquín J García
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Batista APC, da Silva TG, Teixeira ÁA, de Medeiros PL, Teixeira VW, Alves LC, dos Santos FA, Silva EC. Ultrastructural aspects of melatonin cytotoxicity on Caco-2 cells in vitro. Micron 2014; 59:17-23. [DOI: 10.1016/j.micron.2013.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 01/10/2023]
|
46
|
Jardim-Perassi BV, Arbab AS, Ferreira LC, Borin TF, Varma NRS, Iskander ASM, Shankar A, Ali MM, de Campos Zuccari DAP. Effect of melatonin on tumor growth and angiogenesis in xenograft model of breast cancer. PLoS One 2014; 9:e85311. [PMID: 24416386 PMCID: PMC3887041 DOI: 10.1371/journal.pone.0085311] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/26/2013] [Indexed: 12/15/2022] Open
Abstract
As neovascularization is essential for tumor growth and metastasis, controlling angiogenesis is a promising tactic in limiting cancer progression. Melatonin has been studied for their inhibitory properties on angiogenesis in cancer. We performed an in vivo study to evaluate the effects of melatonin treatment on angiogenesis in breast cancer. Cell viability was measured by MTT assay after melatonin treatment in triple-negative breast cancer cells (MDA-MB-231). After, cells were implanted in athymic nude mice and treated with melatonin or vehicle daily, administered intraperitoneally 1 hour before turning the room light off. Volume of the tumors was measured weekly with a digital caliper and at the end of treatments animals underwent single photon emission computed tomography (SPECT) with Technetium-99m tagged vascular endothelial growth factor (VEGF) C to detect in vivo angiogenesis. In addition, expression of pro-angiogenic/growth factors in the tumor extracts was evaluated by membrane antibody array and collected tumor tissues were analyzed with histochemical staining. Melatonin in vitro treatment (1 mM) decreased cell viability (p<0.05). The breast cancer xenografts nude mice treated with melatonin showed reduced tumor size and cell proliferation (Ki-67) compared to control animals after 21 days of treatment (p<0.05). Expression of VEGF receptor 2 decreased significantly in the treated animals compared to that of control when determined by immunohistochemistry (p<0.05) but the changes were not significant on SPECT (p>0.05) images. In addition, there was a decrease of micro-vessel density (Von Willebrand Factor) in melatonin treated mice (p<0.05). However, semiquantitative densitometry analysis of membrane array indicated increased expression of epidermal growth factor receptor and insulin-like growth factor 1 in treated tumors compared to vehicle treated tumors (p<0.05). In conclusion, melatonin treatment showed effectiveness in reducing tumor growth and cell proliferation, as well as in the inhibition of angiogenesis.
Collapse
Affiliation(s)
- Bruna Victorasso Jardim-Perassi
- Department of Biology, Universidade Estadual Paulista, São José do Rio Preto, São Paulo, Brazil
- Laboratório de Investigação Molecular no Câncer, Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Ali S. Arbab
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Lívia Carvalho Ferreira
- Department of Biology, Universidade Estadual Paulista, São José do Rio Preto, São Paulo, Brazil
- Laboratório de Investigação Molecular no Câncer, Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Thaiz Ferraz Borin
- Laboratório de Investigação Molecular no Câncer, Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
- Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Nadimpalli R. S. Varma
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - A. S. M. Iskander
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Adarsh Shankar
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Meser M. Ali
- Cellular and Molecular Imaging Laboratory, Department of Radiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Debora Aparecida Pires de Campos Zuccari
- Laboratório de Investigação Molecular no Câncer, Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
- Department of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
47
|
Update on the role of melatonin in the prevention of cancer tumorigenesis and in the management of cancer correlates, such as sleep-wake and mood disturbances: review and remarks. Aging Clin Exp Res 2013; 25:499-510. [PMID: 24046037 PMCID: PMC3788186 DOI: 10.1007/s40520-013-0118-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/24/2013] [Indexed: 01/24/2023]
Abstract
The aim of this article was to perform a systematic review on the role of melatonin in the prevention of cancer tumorigenesis--in vivo and in vitro--as well as in the management of cancer correlates, such as sleep-wake and mood disturbances. The International Agency for Research on Cancer recently classified "shift-work that involves circadian disruption" as "probably carcinogenic to humans" (Group 2A) based on "limited evidence in humans for the carcinogenicity of shift-work that involves night-work", and "sufficient evidence in experimental animals for the carcinogenicity of light during the daily dark period (biological night)". The clinical implications and the potential uses of melatonin in terms of biologic clock influence (e.g. sleep and mood), immune function, cancer initiation and growth, as well as the correlation between melatonin levels and cancer risk, are hereinafter recorded and summarized. Additionally, this paper includes a description of the newly discovered effects that melatonin has on the management of sleep-wake and mood disturbances as well as with regard to cancer patients' life quality. In cancer patients depression and insomnia are frequent and serious comorbid conditions which definitely require a special attention. The data presented in this review encourage the performance of new clinical trials to investigate the possible use of melatonin in cancer patients suffering from sleep-wake and mood disturbances, also considering that melatonin registered a low toxicity in cancer patients.
Collapse
|
48
|
Liu L, Xu Y, Reiter RJ. Melatonin inhibits the proliferation of human osteosarcoma cell line MG-63. Bone 2013; 55:432-8. [PMID: 23470834 DOI: 10.1016/j.bone.2013.02.021] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 01/03/2013] [Accepted: 02/08/2013] [Indexed: 12/16/2022]
Abstract
It seems established that the onset of osteosarcoma and the reduction in melatonin production run in parallel; this suggests that the decline in the cancer-inhibiting agent, melatonin, may contribute to the occurrence of osteosarcoma and that melatonin supplementation may have promise for preventing the development and progression of this condition. There is, however, no direct evidence regarding an antiproliferative effect of melatonin in osteosarcoma cells. In the current study, we examined whether melatonin inhibits the proliferation of human osteosarcoma cell line MG-63. MTT staining showed that at 4 mM-10 mM concentrations, melatonin significantly reduced the MG-63 cell proliferation in a dose-dependent and time-dependent manner. Flow cytometry documented that 4 mM melatonin significantly increased the fraction of cells in the G(0)/G(1) phase of the cell cycle, while simultaneously reducing the proportion in the S and G(2)/M phases. Western blot and real-time PCR analyses further confirmed that melatonin's inhibitory effect was possibly because of downregulation of cyclin D1 and CDK4, related to the G(1) phase, and of cyclin B1 and CDK1, related to the G(2)/M phase. There was no downregulation of cyclin E, CDK2, and cyclin A, which are related to G(1)/S transition and S phase. These findings provide evidence that melatonin may significantly inhibit human osteosarcoma cell proliferation in a dose-dependent and time-dependent manner and this inhibition involves the downregulation of cyclin D1, CDK4, cyclin B1 and CDK1.
Collapse
Affiliation(s)
- Lifeng Liu
- Department of Orthopaedics, East Hospital, Tongji University School of Medicine, Shanghai, China.
| | | | | |
Collapse
|
49
|
Proietti S, Cucina A, Reiter RJ, Bizzarri M. Molecular mechanisms of melatonin's inhibitory actions on breast cancers. Cell Mol Life Sci 2013; 70:2139-57. [PMID: 23007844 PMCID: PMC11113894 DOI: 10.1007/s00018-012-1161-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 09/04/2012] [Accepted: 09/05/2012] [Indexed: 02/07/2023]
Abstract
Melatonin is involved in many physiological functions and it plays an important role in many pathological processes as well. Melatonin has been shown to reduce the incidence of experimentally induced cancers and can significantly inhibit the growth of some human tumors, namely hormone-dependent cancers. The anticancer effects of melatonin have been observed in breast cancer, both in in vivo with models of chemically induced rat mammary tumors, and in vitro studies on human breast cancer cell lines. Melatonin acts at different physiological levels and its antitumoral properties are supported by a set of complex, different mechanisms of action, involving apoptosis activation, inhibition of proliferation, and cell differentiation.
Collapse
Affiliation(s)
- Sara Proietti
- Department of Clinical and Molecular Medicine, University “La Sapienza”, Rome, Italy
- Department of Surgery “P.Valdoni”, University “La Sapienza”, Rome, Italy
| | - Alessandra Cucina
- Department of Surgery “P.Valdoni”, University “La Sapienza”, Rome, Italy
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX USA
| | - Mariano Bizzarri
- Systems Biology Group Laboratory, Department of Experimental Medicine, University “La Sapienza”, 14-16, Via Antonio Scarpa, Rome, 00161 Italy
| |
Collapse
|
50
|
Alvarez-García V, González A, Alonso-González C, Martínez-Campa C, Cos S. Regulation of vascular endothelial growth factor by melatonin in human breast cancer cells. J Pineal Res 2013; 54:373-80. [PMID: 23013414 DOI: 10.1111/jpi.12007] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 08/10/2012] [Indexed: 11/28/2022]
Abstract
Melatonin exerts oncostatic effects on breast cancer by interfering with the estrogen-signaling pathways. Melatonin reduces estrogen biosynthesis in human breast cancer cells, surrounding fibroblasts and peritumoral endothelial cells by regulating cytokines that influence tumor microenvironment. This hormone also exerts antiangiogenic activity in tumoral tissue. In this work, our objective was to study the role of melatonin on the regulation of the vascular endothelial growth factor (VEGF) in breast cancer cells. To accomplish this, we cocultured human breast cancer cells (MCF-7) with human umbilical vein endothelial cells (HUVECs). VEGF added to the cultures stimulated the proliferation of HUVECs and melatonin (1 mM) counteracted this effect. Melatonin reduced VEGF production and VEGF mRNA expression in MCF-7 cells. MCF-7 cells cocultured with HUVECs stimulated the endothelial cells proliferation and increased VEGF levels in the culture media. Melatonin counteracted both stimulatory effects on HUVECs proliferation and on VEGF protein levels in the coculture media. Conditioned media from MCF-7 cells increased HUVECs proliferation, and this effect was significantly counteracted by anti-VEGF and 1 mM melatonin. All these findings suggest that melatonin may play a role in the paracrine interactions between malignant epithelial cells and proximal endothelial cells through a downregulatory action on VEGF expression in human breast cancer cells, which decrease the levels of VEGF around endothelial cells. Lower levels of VEGF could be important in reducing the number of estrogen-producing cells proximal to malignant cells as well as decreasing tumoral angiogenesis.
Collapse
Affiliation(s)
- Virginia Alvarez-García
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, Spain
| | | | | | | | | |
Collapse
|