1
|
Rani A. RAR-related orphan receptor alpha and the staggerer mice: a fine molecular story. Front Endocrinol (Lausanne) 2024; 14:1300729. [PMID: 38766309 PMCID: PMC11099308 DOI: 10.3389/fendo.2023.1300729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/15/2023] [Indexed: 05/22/2024] Open
Abstract
The retinoic acid-related orphan receptor alpha (RORα) protein first came into the limelight due to a set of staggerer mice, discovered at the Jackson Laboratories in the United States of America by Sidman, Lane, and Dickie (1962) and genetically deciphered by Hamilton et al. in 1996. These staggerer mice exhibited cerebellar defects, an ataxic gait, a stagger along with several other developmental abnormalities, compensatory mechanisms, and, most importantly, a deletion of 160 kilobases (kb), encompassing the RORα ligand binding domain (LBD). The discovery of the staggerer mice and the subsequent discovery of a loss of the LBD within the RORα gene of these mice at the genetic level clearly indicated that RORα's LBD played a crucial role in patterning during embryogenesis. Moreover, a chance study by Roffler-Tarlov and Sidman (1978) noted reduced concentrations of glutamic acid levels in the staggerer mice, indicating a possible role for the essence of a nutritionally balanced diet. The sequential organisation of the building blocks of intact genes, requires the nucleotide bases of deoxyribonucleic acid (DNA): purines and pyrimidines, both of which are synthesized, upon a constant supply of glutamine, an amino acid fortified in a balanced diet and a byproduct of the carbohydrate and lipid metabolic pathways. A nutritionally balanced diet, along with a metabolic "enzymatic machinery" devoid of mutations/aberrations, was essential in the uninterrupted transcription of RORα during embryogenesis. In addition to the above, following translation, a ligand-responsive RORα acts as a "molecular circadian regulator" during embryogenesis and not only is expressed selectively and differentially, but also promotes differential activity depending on the anatomical and pathological site of its expression. RORα is highly expressed in the central nervous system (CNS) and the endocrine organs. Additionally, RORα and the clock genes are core components of the circadian rhythmicity, with the expression of RORα fluctuating in a night-day-night sigmoidal pattern and undoubtedly serves as an endocrine-like, albeit "molecular-circadian regulator". Melatonin, a circadian hormone, along with tri-iodothyronine and some steroid hormones are known to regulate RORα-mediated molecular activity, with each of these hormones themselves being regulated rhythmically by the hypothalamic-pituitary axis (HPA). The HPA regulates the circadian rhythm and cyclical release of hormones, in a self-regulatory feedback loop. Irregular sleep-wake patterns affect circadian rhythmicity and the ability of the immune system to withstand infections. The staggerer mice with their thinner bones, an altered skeletal musculature, an aberrant metabolic profile, the ataxic gait and an underdeveloped cerebellar cortex; exhibited compensatory mechanisms, that not only allowed the survival of the staggerer mice, but also enhanced protection from microbial invasions and resistance to high-fat-diet induced obesity. This review has been compiled in its present form, more than 14 years later after a chromatin immunoprecipitation (ChIP) cloning and sequencing methodology helped me identify signal transducer and activator of transcription 5 (STAT5) target sequences, one of which was mapped to the first intron of the RORα gene. The 599-base-long sequence containing one consensus TTCNNNGAA (TTCN3GAA) gamma-activated sequence (GAS) and five other non-consensus TTN5AA sequences had been identified from the clones isolated from the STAT5 target sites (fragments) in human phytohemagglutinin-activated CD8+ T lymphocytes, during my doctoral studies between 2006 and 2009. Most importantly, preliminary studies noted a unique RORα expression profile, during a time-course study on the ribonucleic acid (RNA), extracted from human phytohemagglutinin (PHA) activated CD8+ T lymphocytes stimulated with interleukin-2 (IL-2). This review mainly focuses on the "staggerer mice" with one of its first roles materialising during embryogenesis, a molecular-endocrine mediated circadian-like regulatory process.
Collapse
Affiliation(s)
- Aradhana Rani
- Medical Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
- Human Resource Development and Management, Indian Institute of Technology (IIT) Kharagpur, West Bengal, India
- Immunology, King’s College London, London, United Kingdom
| |
Collapse
|
2
|
Zhang D, Jia X, Lin D, Ma J. Melatonin and ferroptosis: Mechanisms and therapeutic implications. Biochem Pharmacol 2023; 218:115909. [PMID: 37931663 DOI: 10.1016/j.bcp.2023.115909] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Ferroptosis, a regulated form of cell death, is characterized by iron-dependent lipid peroxidation leading to oxidative damage to cell membranes. Cell sensitivity to ferroptosis is influenced by factors such as iron overload, lipid metabolism, and the regulation of the antioxidant system. Melatonin, with its demonstrated capacity to chelate iron, modulate iron metabolism proteins, regulate lipid peroxidation, and regulate antioxidant systems, has promise as a potential therapeutic agent in mediating ferroptosis. The availability of approved drugs targeting ferroptosis is limited; therefore, melatonin is a candidate for broad application due to its safety and efficacy in attenuating ferroptosis in noncancerous diseases. Melatonin has been demonstrated to attenuate ferroptosis in cellular and animal models of noncancerous diseases, showcasing effectiveness in organs such as the heart, brain, lung, liver, kidney, and bone. This review outlines the molecular mechanisms of ferroptosis, investigates melatonin's potential effects on ferroptosis, and discusses melatonin's therapeutic potential as a promising intervention against diseases associated with ferroptosis. Through this discourse, we aim to lay a strong foundation for developing melatonin as a therapeutic strategy to modulate ferroptosis in a variety of disease contexts.
Collapse
Affiliation(s)
- Dongni Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiaotong Jia
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
3
|
Ashton A, Clark J, Fedo J, Sementilli A, Fragoso YD, McCaffery P. Retinoic Acid Signalling in the Pineal Gland Is Conserved across Mammalian Species and Its Transcriptional Activity Is Inhibited by Melatonin. Cells 2023; 12:286. [PMID: 36672220 PMCID: PMC9856906 DOI: 10.3390/cells12020286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
The pineal gland is integral to the circadian timing system due to its role in nightly melatonin production. Retinoic acid (RA) is a potent regulator of gene transcription and has previously been found to exhibit diurnal changes in synthesis and signalling in the rat pineal gland. This study investigated the potential for the interaction of these two systems. PCR was used to study gene expression in mouse and human pineal glands, ex-vivo organotypic cultured rat pineal gland and cell lines. The mouse and human pineal glands were both found to express the necessary components required for RA signalling. RA influences the circadian clock in the brain, therefore the short-term effect of RA on clock gene expression was determined in ex vivo rat pineal glands but was not found to rapidly regulate Per1, Per2, Bmal1, or Cry1. The interaction between RA and melatonin was also investigated and, unexpectedly, melatonin was found to suppress the induction of gene transcription by RA. This study demonstrates that pineal expression of the RA signalling system is conserved across mammalian species. There is no short-term regulation of the circadian clock but an inhibitory effect of melatonin on RA transcriptional activity was demonstrated, suggesting that there may be functional cross-talk between these systems.
Collapse
Affiliation(s)
- Anna Ashton
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Jason Clark
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Julia Fedo
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Angelo Sementilli
- Department of Physiopathology, Universidade Metropolitana de Santos and Centro, Universitario Lusíada, Santos 11050-071, SP, Brazil
| | - Yara D. Fragoso
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Department of Post Graduate Studies, Universidade Metropolitana de Santos, Santos 11045-002, SP, Brazil
| | - Peter McCaffery
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| |
Collapse
|
4
|
Hagström A, Kal Omar R, Williams PA, Stålhammar G. The rationale for treating uveal melanoma with adjuvant melatonin: a review of the literature. BMC Cancer 2022; 22:398. [PMID: 35413810 PMCID: PMC9006630 DOI: 10.1186/s12885-022-09464-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/28/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Uveal melanoma is a rare form of cancer with high mortality. The incidence of metastases is attributed to early seeding of micrometastases from the eye to distant organs, primarily the liver. Once these seeded clusters of dormant tumor cells grow into larger radiologically detectable macrometastases, median patient survival is about 1 year. Melatonin is an important hormone for synchronizing circadian rhythms. It is also involved in other aspects of human physiology and may offer therapeutic benefits for a variety of diseases including cancer. METHODS Articles involving the physiological effects of melatonin, pharmacokinetics, and previous use in cancer studies were acquired using a comprehensive literature search in the Medline (PubMed) and Web of Science databases. In total, 147 publications were selected and included in the review. RESULTS Melatonin has been observed to suppress the growth of cancer cells, inhibit metastatic spread, enhance immune system functions, and act as an anti-inflammatory in both in vitro and in vivo models. Melatonin may also enhance the efficacy of cancer treatments such as immuno- and chemotherapy. Numerous studies have shown promising results for oral melatonin supplementation in patients with other forms of cancer including cutaneous malignant melanoma. Cell line and animal studies support a hypothesis in which similar benefits may exist for uveal melanoma. CONCLUSIONS Given its low cost, good safety profile, and limited side effects, there may be potential for the use of melatonin as an adjuvant oncostatic treatment. Future avenues of research could include clinical trials to evaluate the effect of melatonin in prevention of macrometastases of uveal melanoma.
Collapse
Affiliation(s)
- Anna Hagström
- Department of Medicine, Karolinska Institutet, D1:04, 171 76, Stockholm, Sweden.
| | - Ruba Kal Omar
- Department of Medicine, Karolinska Institutet, D1:04, 171 76, Stockholm, Sweden.
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64, Stockholm, Sweden
| | - Gustav Stålhammar
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64, Stockholm, Sweden
- St. Erik Eye Hospital, Box 4078, 171 04, Stockholm, Sweden
| |
Collapse
|
5
|
Benna C, Rajendran S, Spiro G, Menin C, Dall'Olmo L, Rossi CR, Mocellin S. Gender-specific associations between polymorphisms of the circadian gene RORA and cutaneous melanoma susceptibility. J Transl Med 2021; 19:57. [PMID: 33549124 PMCID: PMC7866430 DOI: 10.1186/s12967-021-02725-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/28/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Melanoma is the deadliest of skin cancers and has an increasing annual incidence worldwide. It is a multi-factorial disease most likely arising from both genetic predisposition and environmental exposure to ultraviolet light. Genetic variability of the components of the biological circadian clock is recognized to be a risk factor for different type of cancers. Moreover, two variants of a clock gene, RORA, have been associated with melanoma patient's prognosis. Our aim is to test the hypothesis that specific single nucleotide polymorphisms (SNPs) of the circadian clock genes may significantly influence the predisposition to develop cutaneous melanoma or the outcome of melanoma patients. METHODS We genotyped 1239 subjects, 629 cases of melanoma and 610 healthy controls in 14 known SNPs of seven selected clock genes: AANAT, CLOCK, NPAS2, PER1, PER2, RORA, and TIMELESS. Genotyping was conducted by q-PCR. Multivariate logistic regression was employed for susceptibility of melanoma assessment, modeled additively. Subgroup analysis was performed by gender. For the female subgroup, a further discrimination was performed by age. For prognosis of melanoma assessment, multivariate Cox proportional hazard regression was employed. The Benjamini-Hochberg method was utilized as adjustment for multiple comparisons. RESULTS We identified two RORA SNPs statistically significant with respect to the association with melanoma susceptibility. Considering the putative role of RORA as a nuclear steroid hormone receptor, we conducted a subgroup analysis by gender. Interestingly, the RORA rs339972 C allele was associated with a decreased predisposition to develop melanoma only in the female subgroup (OR 0.67; 95% CI 0.51-0.88; P = 0.003) while RORA rs10519097 T allele was associated with a decreased predisposition to develop melanoma only in the male subgroup (OR 0.62; 95% CI 0.44-0.87; P = 0.005). Moreover, the RORA rs339972 C allele had a decreased susceptibility to develop melanoma only in females aged over 50 years old (OR 0.67; 95% CI 0.54-0.83; P = 0.0002). None of the studied SNPs were significantly associated with the prognosis. CONCLUSIONS Overall, we cannot ascertain that circadian pathway genetic variation is involved in melanoma susceptibility or prognosis. Nevertheless, we identified an interesting relationship between melanoma susceptibility and RORA polymorphisms acting in sex-specific manner and which is worth further future investigation.
Collapse
Affiliation(s)
- Clara Benna
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy. .,First Surgical Clinic, Azienda Ospedaliera Padova, Padova, Italy.
| | - Senthilkumar Rajendran
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giovanna Spiro
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Chiara Menin
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology (IOV - IRCCS), Padova, Italy
| | - Luigi Dall'Olmo
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy.,Surgical Oncology Unit, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy
| | - Carlo Riccardo Rossi
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy.,Surgical Oncology Unit, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy
| | - Simone Mocellin
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy.,Surgical Oncology Unit, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy
| |
Collapse
|
6
|
Ladurner A, Schwarz PF, Dirsch VM. Natural products as modulators of retinoic acid receptor-related orphan receptors (RORs). Nat Prod Rep 2021; 38:757-781. [PMID: 33118578 DOI: 10.1039/d0np00047g] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Covering: 1994 to 2020 Retinoic acid receptor-related orphan receptors (RORs) belong to a subfamily of the nuclear receptor superfamily and possess prominent roles in circadian rhythm, metabolism, inflammation, and cancer. They have been subject of research for over two decades and represent attractive but challenging drug targets. Natural products were among the first identified ligands of RORs and continue to be of interest to this day. This review focuses on ligands and indirect modulators of RORs from natural sources and explores their roles in a therapeutic context.
Collapse
Affiliation(s)
- Angela Ladurner
- Department of Pharmacognosy, University of Vienna, Vienna, Austria.
| | - Patrik F Schwarz
- Department of Pharmacognosy, University of Vienna, Vienna, Austria.
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Samanta S. Melatonin: an endogenous miraculous indolamine, fights against cancer progression. J Cancer Res Clin Oncol 2020; 146:1893-1922. [PMID: 32583237 DOI: 10.1007/s00432-020-03292-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Melatonin is an amphipathic indolamine molecule ubiquitously present in all organisms ranging from cyanobacteria to humans. The pineal gland is the site of melatonin synthesis and secretion under the influence of the retinohypothalamic tract. Some extrapineal tissues (skin, lens, gastrointestinal tract, testis, ovary, lymphocytes, and astrocytes) also enable to produce melatonin. Physiologically, melatonin regulates various functions like circadian rhythm, sleep-wake cycle, gonadal activity, redox homeostasis, neuroprotection, immune-modulation, and anticancer effects in the body. Inappropriate melatonin secretion advances the aging process, tumorigenesis, visceral adiposity, etc. METHODS: For the preparation of this review, I had reviewed the literature on the multidimensional activities of melatonin from the NCBI website database PubMed, Springer Nature, Science Direct (Elsevier), Wiley Online ResearchGate, and Google Scholar databases to search relevant articles. Specifically, I focused on the roles and mechanisms of action of melatonin in cancer prevention. RESULTS The actions of melatonin are primarily mediated by G-protein coupled MT1 and MT2 receptors; however, several intracellular protein and nuclear receptors can modulate the activity. Normal levels of the melatonin protect the cells from adverse effects including carcinogenesis. Therapeutically, melatonin has chronomedicinal value; it also shows a remarkable anticancer property. The oncostatic action of melatonin is multidimensional, associated with the advancement of apoptosis, the arrest of the cell cycle, inhibition of metastasis, and antioxidant activity. CONCLUSION The present review has emphasized the mechanism of the anti-neoplastic activity of melatonin that increases the possibilities of the new approaches in cancer therapy.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department Physiology, Midnapore College, Paschim Medinipur, Midnapore, West Bengal, 721101, India.
| |
Collapse
|
8
|
Yan G, Lei H, He M, Gong R, Wang Y, He X, Li G, Pang P, Li X, Yu S, Du W, Yuan Y. Melatonin triggers autophagic cell death by regulating RORC in Hodgkin lymphoma. Biomed Pharmacother 2020; 123:109811. [PMID: 31924597 DOI: 10.1016/j.biopha.2020.109811] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/23/2019] [Accepted: 12/29/2019] [Indexed: 01/07/2023] Open
Abstract
Melatonin (Mel) has been shown to involve in many essential cell functions via modulating many signaling pathways. We for the first time investigated that Mel exerted anti-tumor activities in Hodgkin lymphoma (HL) via inhibiting cell proliferation and promoting cell apoptosis. Further study revealed that Mel treatment increased expression of LC3-II and decreased p62 proteins with the enhanced production of autolysosome, indicating it induced activation of autophagy. Nevertheless, Mel treatment together with autophagy inhibitors 3-MA or CQ exacerbated the damage effect of Mel in HL cells, which means autophagy plays a protective role in this process. Furthermore, we found Mel treatment increased the expression of G protein-coupled receptors MT2 and retinoic acid-related orphan receptors (RORs), eg. RORA, RORB and RORC. While RORC has the highest increase in Mel treated HL cells. In addition, RORC overexpression induced autophagy activation. Therefore, Mel showed tumor-suppressive role due to an increased level of RORC induced autophagy in HL.
Collapse
Affiliation(s)
- Gege Yan
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Hong Lei
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Mingyu He
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Rui Gong
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Yang Wang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Xiaoqi He
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Guanghui Li
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Ping Pang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Xin Li
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Shuting Yu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Weijie Du
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
| | - Ye Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Department of Clinical Pharmarcology, College of Pharmacy, Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
9
|
Effect of the melatonin nuclear receptor RORα on monochromatic light-induced T-lymphocyte proliferation in chicken thymus. Immunol Lett 2019; 213:21-29. [DOI: 10.1016/j.imlet.2019.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/09/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022]
|
10
|
Liu L, Wang T, Yang X, Xu C, Liao Z, Wang X, Su D, Li Y, Zhou H, Qiu X, Chen Y, Huang D, Lian C, Su P. MTNR1B loss promotes chordoma recurrence by abrogating melatonin-mediated β-catenin signaling repression. J Pineal Res 2019; 67:e12588. [PMID: 31140197 DOI: 10.1111/jpi.12588] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/16/2019] [Accepted: 05/22/2019] [Indexed: 01/03/2023]
Abstract
Chordoma is an extremely rare malignant bone tumor with a high rate of relapse. While cancer stem cells (CSCs) are closely associated with tumor recurrence, which depend on its capacity to self-renew and induce chemo-/radioresistance, whether and how CSCs participate in chordoma recurrence remains unclear. The current study found that tumor cells in recurrent chordoma displayed more dedifferentiated CSC-like properties than those in corresponding primary tumor tissues. Meanwhile, MTNR1B deletion along with melatonin receptor 1B (MTNR1B) down-regulation was observed in recurrent chordoma. Further investigation revealed that activation of Gαi2 by MTNR1B upon melatonin stimulation could inhibit SRC kinase activity via recruiting CSK and SRC, increasing SRC Y530 phosphorylation, and decreasing SRC Y419 phosphorylation. This subsequently suppressed β-catenin signaling and stemness via decreasing β-catenin p-Y86/Y333/Y654. However, MTNR1B loss in chordoma mediated increased CSC properties, chemoresistance, and tumor progression by releasing melatonin's repression of β-catenin signaling. Clinically, MTNR1B deletion was found to correlate with patients' survival. Together, our study establishes a novel convergence between melatonin and β-catenin signaling pathways and reveals the significance of this cross talk in chordoma recurrence. Besides, we propose that MTNR1B is a potential biomarker for prediction of chordoma prognosis and selection of treatment options, and chordoma patients might benefit from targeting MTNR1B/Gαi2/SRC/β-catenin axis.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/deficiency
- Biomarkers, Tumor/genetics
- Bone Neoplasms/drug therapy
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Bone Neoplasms/pathology
- Cell Line, Tumor
- Chondroma/drug therapy
- Chondroma/genetics
- Chondroma/metabolism
- Chondroma/pathology
- Female
- Humans
- Melatonin/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Receptor, Melatonin, MT2/deficiency
- Receptor, Melatonin, MT2/metabolism
- Signal Transduction/drug effects
- Xenograft Model Antitumor Assays
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Lei Liu
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Center for Peripheral Nerve Tissue Engineering and Technology Research, Guangzhou, China
- Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tingting Wang
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Yang
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Caixia Xu
- Research Centre for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiheng Liao
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xudong Wang
- Department of Spine Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Deying Su
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongyong Li
- Research Centre for Translational Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hang Zhou
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xianjian Qiu
- Department of Spine Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuyu Chen
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dongsheng Huang
- Department of Spine Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chengjie Lian
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peiqiang Su
- Department of Orthopaedic Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Center for Peripheral Nerve Tissue Engineering and Technology Research, Guangzhou, China
- Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China
| |
Collapse
|
11
|
Dattilo MA, Benzo Y, Herrera LM, Prada JG, Castillo AF, Orlando UD, Podesta EJ, Maloberti PM. Regulatory mechanisms leading to differential Acyl-CoA synthetase 4 expression in breast cancer cells. Sci Rep 2019; 9:10324. [PMID: 31311992 PMCID: PMC6635356 DOI: 10.1038/s41598-019-46776-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023] Open
Abstract
Acyl-CoA synthetase 4 (ACSL4) overexpression plays a causal role in the aggressiveness of triple negative breast cancer. In turn, a negative correlation has been established between ACSL4 and estrogen receptor alpha (ERα) expression. However, the upstream regulatory mechanisms leading to differential ACSL4 expression between triple negative breast cancer and ERα-positive cells remained unknown. We performed the characterization of the human ACSL4 promoter and the identification of transcription factors involved. Deletional analysis demonstrated the proximal 43 base pairs of the promoter are involved in overexpression. By site directed mutagenesis we describe that retinoid-related orphan receptor alpha (RORα), Sp1 and E2F elements are involved in the promoter activity. We established for the first time that estrogen-related receptor alpha (ERRα) is a transcription factor involved in the higher activation of the human ACSL4 promoter in breast cancer cells. Furthermore, a combination of inhibitors of ACSL4 and ERRα produced a synergistic decrease in MDA-MB-231 cell proliferation. We also demonstrated that ERα restoration in triple negative breast cancer cells downregulates ACSL4 expression. The results presented in this manuscript demonstrated transcriptional mechanism is involved in the different expression of ACSL4 in human breast cancer cell lines of different aggressiveness.
Collapse
Affiliation(s)
- Melina A Dattilo
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Yanina Benzo
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Lucía M Herrera
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Jesica G Prada
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Ana F Castillo
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Ulises D Orlando
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Ernesto J Podesta
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina
| | - Paula M Maloberti
- Biomedical Research Institute, INBIOMED, Department of Biochemistry, School of Medicine, University of Buenos Aires, CABA, Buenos Aires, Argentina.
| |
Collapse
|
12
|
Cipolla-Neto J, Amaral FGD. Melatonin as a Hormone: New Physiological and Clinical Insights. Endocr Rev 2018; 39:990-1028. [PMID: 30215696 DOI: 10.1210/er.2018-00084] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
Melatonin is a ubiquitous molecule present in almost every live being from bacteria to humans. In vertebrates, besides being produced in peripheral tissues and acting as an autocrine and paracrine signal, melatonin is centrally synthetized by a neuroendocrine organ, the pineal gland. Independently of the considered species, pineal hormone melatonin is always produced during the night and its production and secretory episode duration are directly dependent on the length of the night. As its production is tightly linked to the light/dark cycle, melatonin main hormonal systemic integrative action is to coordinate behavioral and physiological adaptations to the environmental geophysical day and season. The circadian signal is dependent on its daily production regularity, on the contrast between day and night concentrations, and on specially developed ways of action. During its daily secretory episode, melatonin coordinates the night adaptive physiology through immediate effects and primes the day adaptive responses through prospective effects that will only appear at daytime, when melatonin is absent. Similarly, the annual history of the daily melatonin secretory episode duration primes the central nervous/endocrine system to the seasons to come. Remarkably, maternal melatonin programs the fetuses' behavior and physiology to cope with the environmental light/dark cycle and season after birth. These unique ways of action turn melatonin into a biological time-domain-acting molecule. The present review focuses on the above considerations, proposes a putative classification of clinical melatonin dysfunctions, and discusses general guidelines to the therapeutic use of melatonin.
Collapse
Affiliation(s)
- José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
13
|
Li T, Ni L, Zhao Z, Liu X, Lai Z, Di X, Xie Z, Song X, Wang X, Zhang R, Liu C. Melatonin attenuates smoking-induced hyperglycemia via preserving insulin secretion and hepatic glycogen synthesis in rats. J Pineal Res 2018; 64:e12475. [PMID: 29437243 PMCID: PMC5947659 DOI: 10.1111/jpi.12475] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/02/2018] [Indexed: 12/11/2022]
Abstract
Epidemiology survey indicated that cigarette smoking is a risk factor of diabetes. However, the precise mechanisms remain to be clarified. In this study, we found that smoking caused metabolic malfunctions on pancreas and liver in experimental animal model. These were indicated by hyperglycemia, increased serum hemoglobin A1c level and decreased insulin secretion, inhibition of liver glycogen synthase (LGS), and hepatic glycogen synthesis. Mechanistic studies revealed that all these alterations were caused by the inflammatory reaction and reactive oxygen species (ROS) induced by the smoking. Melatonin treatment significantly preserved the functions of both pancreas and liver by reducing β cell apoptosis, CD68-cell infiltration, ROS production, and caspase-3 expression. The siRNA-knockdown model identified that the protective effects of melatonin were mediated by melatonin receptor-2 (MT2). This study uncovered potentially underlying mechanisms related to the association between smoking and diabetes. In addition, it is, for first time, to report that melatonin effectively protects against smoking-induced glucose metabolic alterations and the signal transduction pathway of melatonin is mainly mediated by its MT2 receptor. These observations provide solid evidence for the clinically use of melatonin to reduce smoking-related diabetes, and the therapeutic regimens are absent currently.
Collapse
Affiliation(s)
- Tianjia Li
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Leng Ni
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhewei Zhao
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinnong Liu
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhichao Lai
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiao Di
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhibo Xie
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xitao Song
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xuebin Wang
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Rui Zhang
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Changwei Liu
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
14
|
RORα2 requires LSD1 to enhance tumor progression in breast cancer. Sci Rep 2017; 7:11994. [PMID: 28931919 PMCID: PMC5607251 DOI: 10.1038/s41598-017-12344-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/07/2017] [Indexed: 11/30/2022] Open
Abstract
Retinoic acid-related orphan receptor α (RORα) regulates diverse physiological processes, including inflammatory responses, lipid metabolism, circadian rhythm, and cancer biology. RORα has four different isoforms which have distinct N-terminal domains but share identical DNA binding domain and ligand binding domain in human. However, lack of specific antibody against each RORα isoform makes biochemical studies on each RORα isoform remain unclear. Here, we generate RORα2-specific antibody and characterize the role of RORα2 in promoting tumor progression in breast cancer. RORα2 requires lysine specific demethylase 1 (LSD1/KDM1A) as a coactivator for transcriptional activation of RORα2 target genes, exemplified by CTNND1. Intriguingly, RORα2 and LSD1 protein levels are dramatically elevated in human breast cancer specimens compared to normal counterparts. Taken together, our studies indicate that LSD1-mediated RORα2 transcriptional activity is important to promote tumor cell migration in human breast cancer as well as breast cancer cell lines. Therefore, our data establish that suppression of LSD1-mediated RORα2 transcriptional activity may be potent therapeutic strategy to attenuate tumor cell migration in human breast cancer.
Collapse
|
15
|
Lo Sardo F, Muti P, Blandino G, Strano S. Melatonin and Hippo Pathway: Is There Existing Cross-Talk? Int J Mol Sci 2017; 18:ijms18091913. [PMID: 28878191 PMCID: PMC5618562 DOI: 10.3390/ijms18091913] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022] Open
Abstract
Melatonin is an indolic hormone that regulates a plethora of functions ranging from the regulation of circadian rhythms and antioxidant properties to the induction and maintenance of tumor suppressor pathways. It binds to specific receptors as well as to some cytosolic proteins, leading to several cellular signaling cascades. Recently, the involvement of melatonin in cancer insurgence and progression has clearly been demonstrated. In this review, we will first describe the structure and functions of melatonin and its receptors, and then discuss both molecular and epidemiological evidence on melatonin anticancer effects. Finally, we will shed light on potential cross-talk between melatonin signaling and the Hippo signaling pathway, along with the possible implications for cancer therapy.
Collapse
Affiliation(s)
- Federica Lo Sardo
- Oncogenomic and Epigenetic Unit, Molecular Chemoprevention Group, Department of Research, Diagnosis and Innovative Technologies, Translational Research Area, Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144 Rome, Italy.
| | - Paola Muti
- Department of Oncology, Juravinski Cancer Center, McMaster University, Hamilton, ON L8S 4L8, Canada.
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, Molecular Chemoprevention Group, Department of Research, Diagnosis and Innovative Technologies, Translational Research Area, Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144 Rome, Italy.
| | - Sabrina Strano
- Oncogenomic and Epigenetic Unit, Molecular Chemoprevention Group, Department of Research, Diagnosis and Innovative Technologies, Translational Research Area, Regina Elena National Cancer Institute, via Elio Chianesi 53, 00144 Rome, Italy.
| |
Collapse
|
16
|
Martínez-Campa C, Menéndez-Menéndez J, Alonso-González C, González A, Álvarez-García V, Cos S. What is known about melatonin, chemotherapy and altered gene expression in breast cancer. Oncol Lett 2017; 13:2003-2014. [PMID: 28454355 PMCID: PMC5403278 DOI: 10.3892/ol.2017.5712] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 11/17/2016] [Indexed: 02/07/2023] Open
Abstract
Melatonin, synthesized in and released from the pineal gland, has been demonstrated by multiple in vivo and in vitro studies to have an oncostatic role in hormone-dependent tumors. Furthermore, several clinical trials point to melatonin as a promising adjuvant molecule to be considered for cancer treatment. In the past few years, evidence of a broader spectrum of action of melatonin as an antitumor agent has arisen; thus, melatonin appears to also have therapeutic effects in several types of hormone-independent cancer, including ovarian, leukemic, pancreatic, gastric and non-small cell lung carcinoma. In the present study, the latest findings regarding melatonin molecular actions when concomitantly administered with either radiotherapy or chemotherapy in cancer were reviewed, with a particular focus on hormone-dependent breast cancer. Finally, the present study discusses which direction should be followed in the next years to definitely clarify whether or not melatonin administration could protect against non-desirable effects (such as altered gene expression and post-translational protein modifications) caused by chemotherapy or radiotherapy treatments. As treatments move towards personalized medicine, comparative gene expression profiling with and without melatonin may be a powerful tool to better understand the antitumor effects of melatonin, the pineal gland hormone.
Collapse
Affiliation(s)
- Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
- Correspondence to: Dr Carlos Martínez-Campa, Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, Av. Cardenal Herrera Oria s/n, 39011 Santander, Spain, E-mail:
| | - Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Virginia Álvarez-García
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot Watt University, EH14 4AS Edinburgh, UK
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| |
Collapse
|
17
|
Hazan I, Hofmann TG, Aqeilan RI. Tumor Suppressor Genes within Common Fragile Sites Are Active Players in the DNA Damage Response. PLoS Genet 2016; 12:e1006436. [PMID: 27977694 PMCID: PMC5157955 DOI: 10.1371/journal.pgen.1006436] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The role of common fragile sites (CFSs) in cancer remains controversial. Two main views dominate the discussion: one suggests that CFS loci are hotspots of genomic instability leading to inactivation of genes encoded within them, while the other view proposes that CFSs are functional units and that loss of the encoded genes confers selective pressure, leading to cancer development. The latter view is supported by emerging evidence showing that expression of a given CFS is associated with genome integrity and that inactivation of CFS-resident tumor suppressor genes leads to dysregulation of the DNA damage response (DDR) and increased genomic instability. These two viewpoints of CFS function are not mutually exclusive but rather coexist; when breaks at CFSs are not repaired accurately, this can lead to deletions by which cells acquire growth advantage because of loss of tumor suppressor activities. Here, we review recent advances linking some CFS gene products with the DDR, genomic instability, and carcinogenesis and discuss how their inactivation might represent a selective advantage for cancer cells.
Collapse
Affiliation(s)
- Idit Hazan
- Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Thomas G. Hofmann
- Cellular Senescence Group, Department of Epigenetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rami I. Aqeilan
- Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, Vermont, United States of America
- * E-mail:
| |
Collapse
|
18
|
Birdsall TC, Rey S, Martin J, Rogers M. Naturopathic Medicine Analysis. Integr Cancer Ther 2016. [DOI: 10.1177/153473540200100214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Timothy C. Birdsall
- Department of Naturopathic Medicine, Cancer Treatment Centers of America at Midwestern, Regional Medical Center, 2520 Elisha Avenue, Zion, IL 60046
| | - Shauna Rey
- Department of Naturopathic Medicine, Cancer Treatment Centers of America at Midwestern, Regional Medical Center, 2520 Elisha Avenue, Zion, IL 60046
| | - Julie Martin
- Department of Naturopathic Medicine, Cancer Treatment Centers of America at Midwestern, Regional Medical Center, 2520 Elisha Avenue, Zion, IL 60046
| | - Michelle Rogers
- Department of Naturopathic Medicine, Cancer Treatment Centers of America at Midwestern, Regional Medical Center, 2520 Elisha Avenue, Zion, IL 60046
| |
Collapse
|
19
|
Boutin JA. Quinone reductase 2 as a promising target of melatonin therapeutic actions. Expert Opin Ther Targets 2015; 20:303-17. [DOI: 10.1517/14728222.2016.1091882] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jean A Boutin
- Institut de Recherches SERVIER, Pole d’Expertise Biotechnologie, Chimie & Biologie, 125, chemin de Ronde, 78290 Croissy-sur-Seine, France
| |
Collapse
|
20
|
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: 204] [Impact Index Per Article: 22.7] [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
|
21
|
Ribas-Latre A, Del Bas JM, Baselga-Escudero L, Casanova E, Arola-Arnal A, Salvadó MJ, Bladé C, Arola L. Dietary proanthocyanidins modulate the rhythm of BMAL1 expression and induce RORα transactivation in HepG2 cells. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.01.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
22
|
Abstract
WWOX is a gene that spans an extremely large chromosomal region. It is derived from within chromosomal band 16q23.2 which is a region with frequent deletions and other alterations in a variety of different cancers. This chromosomal band also contains the FRA16D common fragile site (CFS). CFSs are chromosomal regions found in all individuals which are highly unstable. WWOX has also been demonstrated to function as a tumor suppressor that is involved in the development of many cancers. Two other highly unstable CFSs, FRA3B (3p14.2) and FRA6E (6q26), also span extremely large genes, FHIT and PARK2, respectively, and these two genes are also found to be important tumor suppressors. There are a number of interesting similarities between these three large CFS genes. In spite of the fact that they are derived from some of the most unstable chromosomal regions in the genome, they are found to be highly evolutionarily conserved and the chromosomal region spanning the mouse homologs of both WWOX and FHIT are also CFSs in mice. Many of the other CFSs also span extremely large genes and many of these are very attractive tumor suppressor candidates. WWOX is therefore a member of a very interesting family of very large CFS genes.
Collapse
Affiliation(s)
- Ge Gao
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - David I Smith
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
23
|
Photoperiodic regulation of nuclear melatonin receptor RORα in lymphoid organs of a tropical rodent Funambulus pennanti: Role in seasonal oxidative stress. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 142:141-53. [DOI: 10.1016/j.jphotobiol.2014.11.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 10/16/2014] [Accepted: 11/03/2014] [Indexed: 11/21/2022]
|
24
|
Gao G, Smith DI. Very large common fragile site genes and their potential role in cancer development. Cell Mol Life Sci 2014; 71:4601-15. [PMID: 25300511 PMCID: PMC11113612 DOI: 10.1007/s00018-014-1753-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 10/24/2022]
Abstract
Common fragile sites (CFSs) are large chromosomal regions that are hot-spots for alterations especially within cancer cells. The three most frequently expressed CFS regions (FRA3B, FRA16D and FRA6E) contain genes that span extremely large genomic regions (FHIT, WWOX and PARK2, respectively), and these genes were found to function as important tumor suppressors. Many other CFS regions contain extremely large genes that are also targets of alterations in multiple cancers, but none have yet been demonstrated to function as tumor suppressors. The loss of expression of just FHIT or WWOX has been found to be associated with a worse overall clinical outcome. Studies in different cancers have revealed that some cancers have decreased expression of multiple large CFS genes. This loss of expression could have a profound phenotypic effect on these cells. In this review, we will summarize the known large common fragile site genes and discuss their potential relationship to cancer development.
Collapse
Affiliation(s)
- Ge Gao
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905 USA
| | - David I. Smith
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
25
|
Kim KJ, Choi JS, Kang I, Kim KW, Jeong CH, Jeong JW. Melatonin suppresses tumor progression by reducing angiogenesis stimulated by HIF-1 in a mouse tumor model. J Pineal Res 2013; 54:264-70. [PMID: 22924616 DOI: 10.1111/j.1600-079x.2012.01030.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 07/13/2012] [Indexed: 12/27/2022]
Abstract
The sustained expansion of a tumor mass requires new blood vessel formation to provide rapidly proliferating tumor cells with an adequate supply of oxygen and nutrients. Hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor angiogenesis and growth by regulating the transcription of genes in response to hypoxic stress. This study was designed to investigate the effects of melatonin on tumor growth and angiogenesis, as well as the mechanism underlying the antitumor activities of melatonin. In this study, we show that the administration of melatonin inhibits tumor growth and blocks tumor angiogenesis in mice. Moreover, melatonin diminished the expression of the HIF-1α protein within the tumor mass during tumorigenesis. Our findings suggest that melatonin is a promising anti-angiogenic therapeutic agent targeting HIF-1α in cancer. Considering that HIF-1α is overexpressed in a majority of human cancers, melatonin could offer a potent therapeutic agent for cancer.
Collapse
Affiliation(s)
- Kil-Jung Kim
- Department of Biomedical Science, Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
26
|
Du J, Xu R. RORα, a potential tumor suppressor and therapeutic target of breast cancer. Int J Mol Sci 2012; 13:15755-66. [PMID: 23443091 PMCID: PMC3546659 DOI: 10.3390/ijms131215755] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 11/17/2012] [Accepted: 11/19/2012] [Indexed: 12/29/2022] Open
Abstract
The function of the nuclear receptor (NR) in breast cancer progression has been investigated for decades. The majority of the nuclear receptors have well characterized natural ligands, but a few of them are orphan receptors for which no ligand has been identified. RORα, one member of the retinoid orphan nuclear receptor (ROR) subfamily of orphan receptors, regulates various cellular and pathological activities. RORα is commonly down-regulated and/or hypoactivated in breast cancer compared to normal mammary tissue. Expression of RORα suppresses malignant phenotypes in breast cancer cells, in vitro and in vivo. Activity of RORα can be categorized into the canonical and non-canonical nuclear receptor pathways, which in turn regulate various breast cancer cellular function, including cell proliferation, apoptosis and invasion. This information suggests that RORα is a potent tumor suppressor and a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Jun Du
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; E-Mail:
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; E-Mail:
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY 40536, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-859-323-7889; Fax: +1-859-257-6030
| |
Collapse
|
27
|
Xiang S, Mao L, Duplessis T, Yuan L, Dauchy R, Dauchy E, Blask DE, Frasch T, Hill SM. Oscillation of clock and clock controlled genes induced by serum shock in human breast epithelial and breast cancer cells: regulation by melatonin. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2012; 6:137-50. [PMID: 23012497 PMCID: PMC3448497 DOI: 10.4137/bcbcr.s9673] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This study investigates differences in expression of clock and clock-controlled genes (CCGs) between human breast epithelial and breast cancer cells and breast tumor xenografts in circadian intact rats and examines if the pineal hormone melatonin influences clock gene and CCG expression. Oscillation of clock gene expression was not observed under standard growth conditions in vitro, however, serum shock (50% horse serum for 2 h) induced oscillation of clock gene and CCG expression in MCF-10A cells, which was repressed or disrupted in MCF-7 cells. Melatonin administration following serum shock differentially suppressed or induced clock gene (Bmal1 and Per2) and CCG expression in MCF10A and MCF-7 cells. These studies demonstrate the lack of rhythmic expression of clock genes and CCGs of cells in vitro and that transplantation of breast cancer cells as xenografts into circadian competent hosts re-establishes a circadian rhythm in the peripheral clock genes of tumor cells.
Collapse
Affiliation(s)
- S Xiang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Slominski RM, Reiter RJ, Schlabritz-Loutsevitch N, Ostrom RS, Slominski AT. Melatonin membrane receptors in peripheral tissues: distribution and functions. Mol Cell Endocrinol 2012; 351:152-66. [PMID: 22245784 PMCID: PMC3288509 DOI: 10.1016/j.mce.2012.01.004] [Citation(s) in RCA: 475] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 01/01/2012] [Indexed: 12/15/2022]
Abstract
Many of melatonin's actions are mediated through interaction with the G-protein coupled membrane bound melatonin receptors type 1 and type 2 (MT1 and MT2, respectively) or, indirectly with nuclear orphan receptors from the RORα/RZR family. Melatonin also binds to the quinone reductase II enzyme, previously defined the MT3 receptor. Melatonin receptors are widely distributed in the body; herein we summarize their expression and actions in non-neural tissues. Several controversies still exist regarding, for example, whether melatonin binds the RORα/RZR family. Studies of the peripheral distribution of melatonin receptors are important since they are attractive targets for immunomodulation, regulation of endocrine, reproductive and cardiovascular functions, modulation of skin pigmentation, hair growth, cancerogenesis, and aging. Melatonin receptor agonists and antagonists have an exciting future since they could define multiple mechanisms by which melatonin modulates the complexity of such a wide variety of physiological and pathological processes.
Collapse
Affiliation(s)
- Radomir M. Slominski
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Russel J. Reiter
- Department of Cellular & Structural Biology, UT Health Science Center, San Antonio, TX 78229-3900, United States
| | - Natalia Schlabritz-Loutsevitch
- Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Rennolds S. Ostrom
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Andrzej T. Slominski
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States
- Division of Dermatology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States
- Corresponding author at: Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, 930 Madison Avenue, Suite 5000, Memphis, TN 38163, United States. Tel.: +1 901 448 3741. (A.T. Slominski)
| |
Collapse
|
29
|
Hardeland R, Madrid JA, Tan DX, Reiter RJ. Melatonin, the circadian multioscillator system and health: the need for detailed analyses of peripheral melatonin signaling. J Pineal Res 2012; 52:139-66. [PMID: 22034907 DOI: 10.1111/j.1600-079x.2011.00934.x] [Citation(s) in RCA: 301] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Evidence is accumulating regarding the importance of circadian core oscillators, several associated factors, and melatonin signaling in the maintenance of health. Dysfunction of endogenous clocks, melatonin receptor polymorphisms, age- and disease-associated declines of melatonin likely contribute to numerous diseases including cancer, metabolic syndrome, diabetes type 2, hypertension, and several mood and cognitive disorders. Consequences of gene silencing, overexpression, gene polymorphisms, and deviant expression levels in diseases are summarized. The circadian system is a complex network of central and peripheral oscillators, some of them being relatively independent of the pacemaker, the suprachiasmatic nucleus. Actions of melatonin on peripheral oscillators are poorly understood. Various lines of evidence indicate that these clocks are also influenced or phase-reset by melatonin. This includes phase differences of core oscillator gene expression under impaired melatonin signaling, effects of melatonin and melatonin receptor knockouts on oscillator mRNAs or proteins. Cross-connections between melatonin signaling pathways and oscillator proteins, including associated factors, are discussed in this review. The high complexity of the multioscillator system comprises alternate or parallel oscillators based on orthologs and paralogs of the core components and a high number of associated factors with varying tissue-specific importance, which offers numerous possibilities for interactions with melatonin. It is an aim of this review to stimulate research on melatonin signaling in peripheral tissues. This should not be restricted to primary signal molecules but rather include various secondarily connected pathways and discriminate between direct effects of the pineal indoleamine at the target organ and others mediated by modulation of oscillators.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Germany.
| | | | | | | |
Collapse
|
30
|
Lardone PJ, Guerrero JM, Fernández-Santos JM, Rubio A, Martín-Lacave I, Carrillo-Vico A. Melatonin synthesized by T lymphocytes as a ligand of the retinoic acid-related orphan receptor. J Pineal Res 2011; 51:454-62. [PMID: 21736617 DOI: 10.1111/j.1600-079x.2011.00909.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Melatonin modulates a wide array of physiological events with pleiotropic effects on the immune system. While the relevance of specific melatonin membrane receptors has been well established for several biological functions, retinoic acid-related orphan receptor alpha (RORα) has been suggested as a mediator of nuclear melatonin signalling by results obtained from pharmacological approaches. However, a melatonin-mediated downstream effect cannot be ruled out, and further evidence is needed to support a direct interaction between melatonin and RORα. Here, we show that RORα is mainly located in human Jurkat T-cell nucleus, and it is co-immunoprecipitated with melatonin. Moreover, immunocytochemistry studies confirmed the co-localization of melatonin and RORα. Melatonin promoted a time-dependent decrease in nuclear RORα levels, suggesting a role in the RORα transcriptional activity. Interestingly, RORα acts as a molecular switch implicated in the mutually exclusive generation of Th17 and Treg cells, both involved in the harm/protection balance of immune conditions such as autoimmunity or acute transplant rejection. Therefore, the identification of melatonin as a natural modulator of RORα gives it a tremendous therapeutic potential for a variety of clinical disorders.
Collapse
Affiliation(s)
- Patricia J Lardone
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Department of Medical Biochemistry and Molecular Biology, University of Seville School of Medicine, Seville, Spain
| | | | | | | | | | | |
Collapse
|
31
|
Blask DE, Hill SM, Dauchy RT, Xiang S, Yuan L, Duplessis T, Mao L, Dauchy E, Sauer LA. Circadian regulation of molecular, dietary, and metabolic signaling mechanisms of human breast cancer growth by the nocturnal melatonin signal and the consequences of its disruption by light at night. J Pineal Res 2011; 51:259-69. [PMID: 21605163 PMCID: PMC3162043 DOI: 10.1111/j.1600-079x.2011.00888.x] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
This review article discusses recent work on the melatonin-mediated circadian regulation and integration of molecular, dietary, and metabolic signaling mechanisms involved in human breast cancer growth and the consequences of circadian disruption by exposure to light at night (LAN). The antiproliferative effects of the circadian melatonin signal are mediated through a major mechanism involving the activation of MT(1) melatonin receptors expressed in human breast cancer cell lines and xenografts. In estrogen receptor (ERα+) human breast cancer cells, melatonin suppresses both ERα mRNA expression and estrogen-induced transcriptional activity of the ERα via MT(1) -induced activation of G(αi2) signaling and reduction of 3',5'-cyclic adenosine monophosphate (cAMP) levels. Melatonin also regulates the transactivation of additional members of the steroid hormone/nuclear receptor super-family, enzymes involved in estrogen metabolism, expression/activation of telomerase, and the expression of core clock and clock-related genes. The anti-invasive/anti-metastatic actions of melatonin involve the blockade of p38 phosphorylation and the expression of matrix metalloproteinases. Melatonin also inhibits the growth of human breast cancer xenografts via another critical pathway involving MT(1) -mediated suppression of cAMP leading to blockade of linoleic acid uptake and its metabolism to the mitogenic signaling molecule 13-hydroxyoctadecadienoic acid (13-HODE). Down-regulation of 13-HODE reduces the activation of growth factor pathways supporting cell proliferation and survival. Experimental evidence in rats and humans indicating that LAN-induced circadian disruption of the nocturnal melatonin signal activates human breast cancer growth, metabolism, and signaling provides the strongest mechanistic support, thus far, for population and ecological studies demonstrating elevated breast cancer risk in night shift workers and other individuals increasingly exposed to LAN.
Collapse
Affiliation(s)
- David E Blask
- Laboratory of Chrono-Neuroendocrine Oncology, Tulane University School of Medicine, New Orleans, LA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hill SM, Blask DE, Xiang S, Yuan L, Mao L, Dauchy RT, Dauchy EM, Frasch T, Duplesis T. Melatonin and associated signaling pathways that control normal breast epithelium and breast cancer. J Mammary Gland Biol Neoplasia 2011; 16:235-45. [PMID: 21773809 DOI: 10.1007/s10911-011-9222-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 06/23/2011] [Indexed: 11/26/2022] Open
Abstract
This review article discusses recent work on the melatonin-mediated circadian regulation and integration of molecular and metabolic signaling mechanisms involved in human breast cancer growth and the associated consequences of circadian disruption by exposure to light-at-night (LAN). The anti-proliferative effects of the circadian melatonin signal are, in general, mediated through mechanisms involving the activation of MT(1) melatonin receptors expressed in human breast cancer cell lines and xenografts. In estrogen receptor-positive (ERα+) human breast cancer cells, melatonin suppresses both ERα mRNA expression and estrogen-induced transcriptional activity of the ERα via MT(1)-induced activation of G(αi2) signaling and reduction of cAMP levels. Melatonin also regulates the transcriptional activity of additional members of the nuclear receptor super-family, enzymes involved in estrogen metabolism, and the expression of core clock and clock-related genes. The anti-invasive/anti-metastatic actions of melatonin involve the blockade of p38 phosphorylation and matrix metalloproteinase expression. Melatonin also inhibits the growth of human breast cancer xenografts via MT(1)-mediated suppression of cAMP leading to a blockade of linoleic acid (LA) uptake and its metabolism to the mitogenic signaling molecule 13-hydroxyoctadecadienoic acid (13-HODE). Down-regulation of 13-HODE reduces the activation of growth factor pathways supporting cell proliferation and survival. Finally, studies in both rats and humans indicate that light-at-night (LAN) induced circadian disruption of the nocturnal melatonin signal activates human breast cancer growth, metabolism, and signaling, providing the strongest mechanistic support, thus far, for epidemiological studies demonstrating the elevated breast cancer risk in night shift workers and other individuals increasingly exposed to LAN.
Collapse
Affiliation(s)
- Steven M Hill
- Department of Structural and Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, LA 70112, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Riggins RB, Mazzotta MM, Maniya OZ, Clarke R. Orphan nuclear receptors in breast cancer pathogenesis and therapeutic response. Endocr Relat Cancer 2010; 17:R213-31. [PMID: 20576803 PMCID: PMC3518023 DOI: 10.1677/erc-10-0058] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors comprise a large family of highly conserved transcription factors that regulate many key processes in normal and neoplastic tissues. Most nuclear receptors share a common, highly conserved domain structure that includes a carboxy-terminal ligand-binding domain. However, a subgroup of this gene family is known as the orphan nuclear receptors because to date there are no known natural ligands that regulate their activity. Many of the 25 nuclear receptors classified as orphan play critical roles in embryonic development, metabolism, and the regulation of circadian rhythm. Here, we review the emerging role(s) of orphan nuclear receptors in breast cancer, with a particular focus on two of the estrogen-related receptors (ERRalpha and ERRgamma) and several others implicated in clinical outcome and response or resistance to cytotoxic or endocrine therapies, including the chicken ovalbumin upstream promoter transcription factors, nerve growth factor-induced B, DAX-1, liver receptor homolog-1, and retinoic acid-related orphan receptor alpha. We also propose that a clearer understanding of the function of orphan nuclear receptors in mammary gland development and normal mammary tissues could significantly improve our ability to diagnose, treat, and prevent breast cancer.
Collapse
Affiliation(s)
- Rebecca B. Riggins
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Mary M. Mazzotta
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Omar Z. Maniya
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Robert Clarke
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
- Department of Physiology and Biophysics, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| |
Collapse
|
34
|
Hill SM, Frasch T, Shulin Xiang, Lin Yuan, Duplessis T, Lulu Mao. Molecular Mechanisms of Melatonin Anticancer Effects. Integr Cancer Ther 2009; 8:337-46. [DOI: 10.1177/1534735409353332] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The authors have shown that, via activation of its MT1 receptor, melatonin modulates the transcriptional activity of various nuclear receptors and the proliferation of both ERα+ and ERα- human breast cancer cells. Employing dominant-negative (DN) and dominant-positive (DP) G proteins, it was demonstrated that Gα i2 proteins mediate the suppression of estrogen-induced ERα transcriptional activity by melatonin, whereas the Gαq proteins mediate the enhancement of retinoid-induced RARα transcriptional activity by melatonin. In primary human breast tumors, the authors’ studies demonstrate an inverse correlation between ERα and MT1 receptor expression, and confocal microscopic studies demonstrate that the MT1I receptor is localized to the caveoli and that its expression can be repressed by estrogen and melatonin. Melatonin, via activation of its MT1 receptor, suppresses the development and growth of breast cancer by regulation of growth factors, regulation of gene expression, regulation of clock genes, inhibition of tumor cell invasion and metastasis, and even regulation of mammary gland development. The authors have previously reported that the clock gene, Period 2 ( Per2), is not expressed in human breast cancer cells but that its reexpression in breast cancer cells results in increased expression of p53 and induction of apoptosis. The authors demonstrate that melatonin, via repression of RORα transcriptional activity, blocks the expression of the clock gene BMAL1 . Melatonin’s blockade of BMAL1 expression is associated with the decreased expression of SIRT1, a member of the Silencing Information Regulator family and a histone and protein deacetylase that inhibits the expression of DNA repair enzymes (p53, BRCA1 & 2, and Ku70) and the expression of apoptosis-associated genes. Finally, the authors developed an MMTV-MT1-flag mammary knock-in transgenic mouse that displays reduced ductal branching, ductal epithelium proliferation, and reduced terminal end bud formation during puberty and pregnancy. Lactating female MT1 transgenic mice show a dramatic reduction in the expression of β-casein and whey acidic milk proteins. Further analyses showed significantly reduced ERα expression in mammary glands of MT1 transgenic mice. These results demonstrate that the MT1 receptor is a major transducer of melatonin’s actions in the breast, suppressing mammary gland development and mediating the anticancer actions of melatonin through multiple pathways.
Collapse
Affiliation(s)
| | | | | | - Lin Yuan
- Tulane University, New Orleans, LA, USA
| | | | - Lulu Mao
- Tulane University, New Orleans, LA, USA
| |
Collapse
|
35
|
Girgert R, Hanf V, Emons G, Gründker C. Membrane-bound melatonin receptor MT1 down-regulates estrogen responsive genes in breast cancer cells. J Pineal Res 2009; 47:23-31. [PMID: 19522736 DOI: 10.1111/j.1600-079x.2009.00684.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Melatonin possesses anti-estrogenic effects on estrogen receptor expressing (ER+) breast cancer cells in culture by reducing cell cycle progression and cell proliferation. There is increasing agreement that on a cellular level the effects of melatonin are primarily induced by the membrane-bound receptor MT1. The participation of a second, nuclear receptor of the group of ligand-dependent transcription factors, called RZRalpha, is under debate. In this study we used a number of breast cancer cell lines differing in their expression of the estrogen receptor and the two known melatonin receptors. In MCF-7 breast cancer cells transfected with a vector carrying the MT1 gene (MCF-7Mel1a) binding of CREB-protein to the cAMP-responsive element of the breast cancer suppressing gene BRCA-1 was more strongly reduced by treatment with melatonin than in the parental cells. Expression of estrogen responsive genes was determined in serum-starved cells, cells stimulated for 16 hr with estradiol and cells subsequently treated with melatonin. Expression of BRCA-1, p53, p21(WAF) and c-myc were up-regulated by estradiol. Treatment of the stimulated cells with melatonin counteracted the increase induced by estradiol almost completely. The more MT1 a cell line expressed, the stronger was the reduction of the expression of the estradiol-induced genes. There was no correlation between the expression of the nuclear receptor RZRalpha and the effects of melatonin on these genes.
Collapse
MESH Headings
- BRCA1 Protein/biosynthesis
- BRCA1 Protein/genetics
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Growth Processes/drug effects
- Cell Growth Processes/genetics
- Cell Line, Tumor
- Cyclin-Dependent Kinase Inhibitor p21/biosynthesis
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- Down-Regulation
- Estradiol/pharmacology
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Melatonin/metabolism
- Melatonin/pharmacology
- Proto-Oncogene Proteins c-myc/biosynthesis
- Proto-Oncogene Proteins c-myc/genetics
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT1/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Estrogen/biosynthesis
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Suppressor Protein p53/biosynthesis
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Rainer Girgert
- Department of Obstetrics and Gynecology, University of Göttingen, Göttingen, Germany
| | | | | | | |
Collapse
|
36
|
Odawara H, Iwasaki T, Horiguchi J, Rokutanda N, Hirooka K, Miyazaki W, Koibuchi Y, Shimokawa N, Iino Y, Takeyoshi I, Koibuchi N. Activation of aromatase expression by retinoic acid receptor-related orphan receptor (ROR) alpha in breast cancer cells: identification of a novel ROR response element. J Biol Chem 2009; 284:17711-9. [PMID: 19439415 PMCID: PMC2719410 DOI: 10.1074/jbc.m109.009241] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 04/30/2009] [Indexed: 11/06/2022] Open
Abstract
Estrogen is a key regulator of the proliferation and differentiation of breast cancer cells. In addition to the estrogen supply from the ovary, estrogen is produced locally from androgen by aromatase. However, the regulation of aromatase gene expression in breast cancer has not yet been fully clarified. Retinoic acid receptor-related orphan receptor (ROR) alpha plays an important role in the differentiation of many organs by regulating the transcription of target genes. Because aromatase and RORalpha are expressed in breast cancer, the effect of RORalpha on aromatase gene expression was studied. RORalpha significantly augmented the expression of aromatase mRNA, particularly those containing exon I.4, in MCF7 cells, and aromatase activities in T47D and MCF7 cells. RORalpha also stimulated the proliferation of these cells. Transient transfection-based reporter gene assays using the promoter at exon I.4 showed that RORalpha augmented the transcription. A series of truncated mutation studies revealed that RORalpha activated the transcription through -147 to +14 bp of the promoter I.4. Furthermore, RORalpha bound to the fragment containing -119 to -107 bp of the promoter in vitro, indicating that this region may contain a novel ROR response element. Chromatin immunoprecipitation assay showed that RORalpha bound to the region containing this site of the promoter I.4 in MCF7 cells. Moreover, we examined clinical samples and found a correlation between RORalpha and aromatase expression. These results suggest that RORalpha directly activates the aromatase expression to accelerate the local production of estrogen, which results in the proliferation of breast cancer cells.
Collapse
MESH Headings
- Aromatase/genetics
- Aromatase/metabolism
- Binding Sites
- Blotting, Western
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Proliferation
- Chromatin Immunoprecipitation
- Electrophoretic Mobility Shift Assay
- Gene Expression Regulation, Neoplastic/physiology
- Humans
- Luciferases
- Mutation/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 1
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Response Elements/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Hiroki Odawara
- From the Departments of Thoracic and Visceral Organ Surgery
| | | | - Jun Horiguchi
- From the Departments of Thoracic and Visceral Organ Surgery
| | - Nana Rokutanda
- From the Departments of Thoracic and Visceral Organ Surgery
| | | | | | - Yukio Koibuchi
- From the Departments of Thoracic and Visceral Organ Surgery
| | | | - Yuichi Iino
- Emergency Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | | | | |
Collapse
|
37
|
Perrone S, Turrisi G, Buonocore G. Antioxidant therapy and neuroprotection in the newborn. ACTA ACUST UNITED AC 2008. [DOI: 10.2217/17455111.2.6.715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Injury to the perinatal brain is a leading cause of childhood mortality and lifelong disability. Despite recent improvements in neonatal care, no effective treatment for perinatal brain lesions is available. The newborn, especially if preterm, is highly prone to oxidative stress (OS) and to the toxic effect of free radicals (FRs). At birth, the newborn is exposed to a relatively hyperoxic environment caused by an increased oxygen bioavailability with greatly enhanced generation of FRs. Additional sources (e.g., inflammation, hypoxia, ischemia, glutamate and free iron release) occur, magnifying OS. In the preterm baby, the perinatal transition is accompanied by the immaturity of the antioxidant systems and the reduced ability to induce efficient homeostatic mechanisms designed to control overproduction of cell-damaging FRs. Improved understanding of the pathophysiological mechanism involved in perinatal brain lesions helps to identify potential targets for neuroprotective interventions, and the knowledge of these mechanisms has enabled scientists to develop new therapeutic strategies that have confirmed their neuroprotective effects in animal studies. Considering the growing role of OS in preterm newborn morbidity in respect to the higher risk of FR damage in these babies, erythropoietin, allopurinol, melatonin and hypothermia demonstrate great promise as potential neuroprotectans. This article provides an overview of the pathogenesis of FR-mediated diseases of the newborn and the antioxidant strategies now tested in order to reduce OS and its damaging effects.
Collapse
Affiliation(s)
| | | | - Giuseppe Buonocore
- Professor of Paediatrics, Department of Pediatrics, Obstetrics & Reproductive Medicine, University of Siena, Italy
| |
Collapse
|
38
|
Hung MW, Tipoe GL, Poon AMS, Reiter RJ, Fung ML. Protective effect of melatonin against hippocampal injury of rats with intermittent hypoxia. J Pineal Res 2008; 44:214-21. [PMID: 18289174 DOI: 10.1111/j.1600-079x.2007.00514.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obstructive sleep apnea (OSA) patients suffer from intermittent hypoxia (IH) and neuropsychologic impairments. Oxidative stress is involved in the pathogenesis of OSA, so the application of an antioxidant may be useful. We evaluated the hypothesis that melatonin would reduce IH-induced hippocampal injury via an increased expression of antioxidant enzymes. Adult Sprague-Dawley rats that had received a daily injection of melatonin or vehicle were exposed to IH for 8 hr/day for 7 or 14 days. The serum and hippocampus were harvested for the measurement of malondialdehyde (MDA). Apoptotic cell death was studied histologically in hippocampal sections. The mRNA expression of inflammatory mediators including tumor necrosis factor-alpha, inducible nitric oxide synthase, cyclooxygenase-2 and antioxidant enzymes including glutathione peroxidase, catalase and copper/zinc superoxide dismutase were examined in the hippocampus by RT-PCR. The results show significant increases in levels of serum and hippocampal MDA, apoptotic cell death and mRNA levels of inflammatory mediators in hypoxic rats when compared with the normoxic controls. Also, mRNA levels of the antioxidant enzymes were decreased in hypoxic animals. In the melatonin-treated hypoxic rats, serum MDA levels were comparable with those in normoxic control rats. Also, melatonin treatment significantly reduced hippocampal MDA levels and totally prevented apoptosis. Moreover, there were a decreased expression of the inflammatory mediators and an elevated expression of antioxidant enzymes in the melatonin injected rats when compared with vehicle-treated animals. These results indicate that melatonin mitigates oxidative stress and the pathogenesis of IH-induced hippocampal injury via its antioxidant and anti-inflammatory properties which includes stimulation of transcriptional regulation of antioxidant enzymes.
Collapse
Affiliation(s)
- Ming-Wai Hung
- Department of Physiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
39
|
García-Navarro A, González-Puga C, Escames G, López LC, López A, López-Cantarero M, Camacho E, Espinosa A, Gallo MA, Acuña-Castroviejo D. Cellular mechanisms involved in the melatonin inhibition of HT-29 human colon cancer cell proliferation in culture. J Pineal Res 2007; 43:195-205. [PMID: 17645698 DOI: 10.1111/j.1600-079x.2007.00463.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The antiproliferative and proapoptotic properties of melatonin in human colon cancer cells in culture were recently reported. To address the mechanisms involved in these actions, HT-29 human colon cancer cells were cultured in RPMI 1640 medium supplemented with fetal bovine serum at 37 degrees C. Cell proliferation was assessed by the incorporation of [(3)H]-thymidine into DNA. Cyclic nucleotide levels, nitrite concentration, glutathione peroxidase and reductase activities, and glutathione levels were assessed after the incubation of these cells with the following drugs: melatonin membrane receptor agonists 2-iodo-melatonin, 2-iodo-N-butanoyl-5-methoxytryptamine, 5-methoxycarbonylamino-N-acetyltryptamine (GR-135,531), and the antagonists luzindole, 4-phenyl-2-propionamidotetralin, and prazosin; the melatonin nuclear receptor agonist CGP 52608, and four synthetic kynurenines analogs to melatonin 2-acetamide-4-(3-methoxyphenyl)-4-oxobutyric acid, 2-acetamide-4-(2-amino-5-methoxyphenyl)-4-oxobutyric acid, 2-butyramide-4-(3-methoxyphenyl)-4-oxobutyric acid and 2-butyramide-4-(2-amino-5-methoxyphenyl)-4-oxobutyric acid. The results show that the membrane receptors are not necessary for the antiproliferative effect of melatonin and the participation of the nuclear receptor in this effect is suggested. Moreover, the antioxidative and anti-inflammatory actions of melatonin, counteracting the oxidative status and reducing the production of nitric oxide by cultured HT-29 cells seem to be directly involved in the oncostatic properties of melatonin. Some of the synthetic kynurenines exert higher antiproliferative effects than melatonin. The results reinforce the clinical interest of melatonin due to the different mechanisms involved in its oncostatic role, and suggest a new synthetic pathway to obtain melatonin agonists with clinical applications to oncology.
Collapse
Affiliation(s)
- Ana García-Navarro
- Departamento de Cirugía, Hospital Universitario San Cecilio, Granada, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Srinivasan V, Spence DW, Pandi-Perumal SR, Trakht I, Esquifino AI, Cardinali DP, Maestroni GJ. Melatonin, environmental light, and breast cancer. Breast Cancer Res Treat 2007; 108:339-50. [PMID: 17541739 DOI: 10.1007/s10549-007-9617-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Accepted: 05/07/2007] [Indexed: 12/15/2022]
Abstract
Although many factors have been suggested as causes for breast cancer, the increased incidence of the disease seen in women working in night shifts led to the hypothesis that the suppression of melatonin by light or melatonin deficiency plays a major role in cancer development. Studies on the 7,12-dimethylbenz[a]anthracene and N-methyl-N-nitrosourea experimental models of human breast cancer indicate that melatonin is effective in reducing cancer development. In vitro studies in MCF-7 human breast cancer cell line have shown that melatonin exerts its anticarcinogenic actions through a variety of mechanisms, and that it is most effective in estrogen receptor (ER) alpha-positive breast cancer cells. Melatonin suppresses ER gene, modulates several estrogen dependent regulatory proteins and pro-oncogenes, inhibits cell proliferation, and impairs the metastatic capacity of MCF-7 human breast cancer cells. The anticarcinogenic action on MCF-7 cells has been demonstrated at the physiological concentrations of melatonin attained at night, suggesting thereby that melatonin acts like an endogenous antiestrogen. Melatonin also decreases the formation of estrogens from androgens via aromatase inhibition. Circulating melatonin levels are abnormally low in ER-positive breast cancer patients thereby supporting the melatonin hypothesis for breast cancer in shift working women. It has been postulated that enhanced endogenous melatonin secretion is responsible for the beneficial effects of meditation as a form of psychosocial intervention that helps breast cancer patients.
Collapse
Affiliation(s)
- V Srinivasan
- Department of Physiology, School of Medical Sciences, University Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | | | | | | | | | | | | |
Collapse
|
41
|
Bartsch C, Bartsch H. The Anti-tumor Activity of Pineal Melatonin and Cancer Enhancing Life Styles in Industrialized Societies. Cancer Causes Control 2006; 17:559-71. [PMID: 16596311 DOI: 10.1007/s10552-005-9011-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This review discusses the potential role of the anti-tumor activity of pineal melatonin for the aetiology and prevention of cancers related to life-styles in industrialized societies, e.g. frequent long-distance flights as well as chronic night shift work leading to circadian disturbances of neuroendocrine parameters including melatonin. Experimental studies show that melatonin controls not only the growth of well-differentiated cancers, but also possesses anti-carcinogenic properties. Therefore, it is plausible that disturbances of circadian melatonin rhythmicity could be functionally involved in elevated cancer risks among aircrew members and nurses frequently working on night shifts. Due to the suppression of melatonin by light it can be assumed that too much artificial light at night could, at least in part, be responsible for generally increasing rates of e.g. breast cancer in industrialized countries. It is discussed under which conditions a transient substitutional therapy with melatonin could be justified or which forms of living could help to physiologically foster melatonin secretion to optimise control over cancerous growth and development.
Collapse
Affiliation(s)
- Christian Bartsch
- Center for Research in Medical and Natural Sciences (MNF), University of Tübingen, ob dem Himmelreich 7, D-72074, Tübingen, Germany.
| | | |
Collapse
|
42
|
Melancon K, Cheng Q, Kiefer TL, Dai J, Lai L, Dong C, Yuan L, Collins A, Thiyagarajah A, Long S, Hill SM. Regression of NMU-induced mammary tumors with the combination of melatonin and 9-cis-retinoic acid. Cancer Lett 2005; 227:39-48. [PMID: 16051031 DOI: 10.1016/j.canlet.2005.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Accepted: 01/07/2005] [Indexed: 10/25/2022]
Abstract
A significant increase in tumor regression was induced in N-nitroso-N-methylurea-induced mammary tumors in rats treated with the combination of melatonin and 9-cis-retinoic acid (9cRA). Treatment groups included: control (ethanolic saline), 9cRA (30 mg/kg chow/day), melatonin 500 microg/day, melatonin 1000 microg/day, melatonin 500 microg/day+9cRA and melatonin 1000 microg/day+9cRA. Rats treated with the lower dose of melatonin 500 microg+9cRA show the greatest degree of tumor regression (78%), with 54% undergoing complete regression and a significant increase in apoptotic cells observed by TUNEL Assay. Furthermore, tumor multiplicity and burden were significantly decreased by the combination of melatonin and 9cRA.
Collapse
Affiliation(s)
- K Melancon
- Department of Transplantation and Surgery, Mayo Clinic, Rochester, MN 555905, USA; Department of Surgery, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tomás-Zapico C, Coto-Montes A. A proposed mechanism to explain the stimulatory effect of melatonin on antioxidative enzymes. J Pineal Res 2005; 39:99-104. [PMID: 16098085 DOI: 10.1111/j.1600-079x.2005.00248.x] [Citation(s) in RCA: 366] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Melatonin, the main secretory product of the pineal gland, is known to collaborate against oxidative stress within cells, but its mechanism of action in terms of stimulating antioxidant enzymes remains unclear. Herein, we propose that melatonin modulates antioxidant enzyme activities via its interaction with calmodulin, which in turn inhibits downstream processes that lead to the inactivation of nuclear RORalpha melatonin receptor. Eventually, this nuclear transcription factor downregulates NF-kappaB-induced antioxidant enzyme expression. Therefore, the increment in antioxidant enzyme activities induced by melatonin involves the inhibition of the RORalpha pathway. Thus, in addition to its direct free radical scavenging activities, melatonin has important actions in oxidative defense by stimulating enzymes which metabolize free radicals and radical products to innocuous metabolites.
Collapse
Affiliation(s)
- Cristina Tomás-Zapico
- Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de Oviedo, Oviedo, Spain
| | | |
Collapse
|
44
|
Tomás-Zapico C, Antonio Boga J, Caballero B, Vega-Naredo I, Sierra V, Alvarez-García O, Tolivia D, Josefa Rodríguez-Colunga M, Coto-Montes A. Coexpression of MT1 and RORalpha1 melatonin receptors in the Syrian hamster Harderian gland. J Pineal Res 2005; 39:21-6. [PMID: 15978053 DOI: 10.1111/j.1600-079x.2005.00210.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Melatonin acts through several specific receptors, including membrane receptors (MT(1) and MT(2)) and members of the RZR/ROR nuclear receptors family, which have been identified in a large variety of mammalian and nonmammalian cells types. Both membrane and nuclear melatonin receptors have been partially characterized in Harderian gland of the Syrian hamster. Nevertheless, the identities of these receptors were unknown until this study, where the coexistence of MT(1) and RORalpha(1) in this gland was determined by nested RT-PCR followed by amplicon sequencing and Western-blot. Furthermore, the cellular localization of both receptors was determined by immunohistochemistry. Thus, MT(1) receptor was localized exclusively at the basal side of the cell acini, supporting the hypothesis that this receptor is activated by the pineal-synthesized melatonin. On the contrary, although a RORalpha(1)-immunoreactivity was observed in nuclei of epithelial cells of both sexes, an extranuclear specific staining, which was more frequently among those cells of males, was also seen. The implication of this possible nuclear exclusion of RORalpha(1) on the role of this indoleamine against oxidative stress is discussed.
Collapse
|
45
|
Abstract
For decades, the important physiological roles of the pineal hormone have inspired scientific investigations. Research efforts have generated a broad amount of information relevant to various genetic aspects of melatonin biology. Nevertheless, our understanding of the effect of genetic factors upon melatonin biosynthesis and the mechanisms of gene expression regulation by melatonin in target tissues is far from complete. The present review makes an effort to summarize and systematize the existing information on the subject, sequentially discussing (i) the effect of genetic factors upon melatonin biosynthesis, (ii) melatonin receptor expression profiles, and (iii) the effect of melatonin upon expression of genes in target tissues.
Collapse
Affiliation(s)
- Sergey V Anisimov
- Section for Neuronal Survival, Wallenberg Neuroscience Center, Department of Physiological Sciences, Lund University, Lund, Sweden.
| | | |
Collapse
|
46
|
Karasek M, Gruszka A, Lawnicka H, Kunert-Radek J, Pawlikowski M. Melatonin inhibits growth of diethylstilbestrol-induced prolactin-secreting pituitary tumor in vitro: possible involvement of nuclear RZR/ROR receptors. J Pineal Res 2003; 34:294-6. [PMID: 12662353 DOI: 10.1034/j.1600-079x.2003.00046.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Melatonin exerts a marked antiproliferative action in numerous experimentally-induced tumors in vivo as well as in both animal and human cell lines in vitro. However, the mechanisms of oncostatic action of melatonin is not clear, and the involvement of both membrane and nuclear receptors are suggested. Therefore, the aim of this study was to investigate effects of melatonin, and both agonist (CGP 52608), and antagonist (CGP 55644) of RZR/ROR nuclear receptors on the growth of diethylstilbestrol-induced rat prolactin-secreting pituitary tumor cells in vitro. Pituitary tumors were induced by subcutaneous implantation of a single silastic capsule containing 10 mg of diethylstilbestrol in 4-wk-old male Fischer 344 rats. Four months after the implantation of capsules the animals were killed by decapitation, pituitary tumors were aseptically removed, mechanically dispersed, and enzymatically digested with 0.2% collagenase and 0.2% hyaluronidase. The cells (6 x 105 cells/well) were incubated for 24 hr in the presence of melatonin, CGP 52608, CGP 55644 and CGP 55644 plus melatonin (at the concentrations of 107 and 10-9 m) at 37 degrees C in the humidified atmosphere of 95% air and 5% CO2. The group with the addition of solvent only served as control. The growth of cell was measured using the EZ4U system. Statistical analysis was performed using ANOVA followed by LSD test. Both melatonin and CGP 52608 significantly suppressed growth of tumor cells in vitro in both used concentrations. CGP 55644 stimulated growth of tumor cells and blocked the inhibitory effects of melatonin in vitro. Results of the present study as well as other experimental evidence strongly support the hypothesis that both membrane and nuclear receptors are involved in the oncostatic action of melatonin, and indicate that nuclear signalling plays an important role in this process.
Collapse
Affiliation(s)
- Michal Karasek
- Department of Electron Microscopy, Chair of Pathomorphology, Medical University of Lodz, Lodz, Poland.
| | | | | | | | | |
Collapse
|
47
|
Klosen P, Bienvenu C, Demarteau O, Dardente H, Guerrero H, Pévet P, Masson-Pévet M. The mt1 melatonin receptor and RORbeta receptor are co-localized in specific TSH-immunoreactive cells in the pars tuberalis of the rat pituitary. J Histochem Cytochem 2002; 50:1647-57. [PMID: 12486087 DOI: 10.1177/002215540205001209] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The pars tuberalis (PT) of the pituitary represents an important target site for the time-pacing pineal hormone melatonin because it expresses a large number of mt1 receptors. Functional studies suggest that the PT mediates the seasonal effects of melatonin on prolactin (PRL) secretion. The aim of this study was the characterization of the phenotype of melatonin-responsive cells. Furthermore, we determined whether RORbeta, a retinoid orphan receptor present in the PT, was co-expressed in the same cells. We combined nonradioactive in situ hybridization (ISH) with hapten-labeled riboprobes for detection of the receptors and immunocytochemistry (ICC) for detection of alphaGSU (alpha-glycoprotein subunit), betaTSH, betaFSH, betaLH, GH, PRL, and ACTH. Expression of mt1 mRNA was found in small round cells, co-localized with alphaGSU and betaTSH. However, not all betaTSH-containing cells expressed mt1 mRNA. The distribution of mt1- and RORbeta-positive cells appeared to overlap, although more cells were labeled for RORbeta than for mt1. Gonadotrophs, as well as other pars distalis cell types, were never labeled for mt1 melatonin receptor. Therefore, this study identifies the "specific" cells of the PT as the mt1 melatonin receptor-expressing cells.
Collapse
Affiliation(s)
- Paul Klosen
- Neurobiologie des Rythmes, CNRS-UMR 7518, IFR 37, Université Louis Pasteur, Strasbourg, France.
| | | | | | | | | | | | | |
Collapse
|
48
|
Ram PT, Dai J, Yuan L, Dong C, Kiefer TL, Lai L, Hill SM. Involvement of the mt1 melatonin receptor in human breast cancer. Cancer Lett 2002; 179:141-50. [PMID: 11888668 DOI: 10.1016/s0304-3835(01)00873-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Two putative melatonin receptors have been described including the cell surface G-protein-linked receptors, mt1 and MT2, and the nuclear retinoic orphan receptor alpha (RORalpha). The mt1 receptor, but not the MT2 receptor, is expressed in human breast tumor cell lines, and melatonin-induced growth suppression can be mimicked by the mt1 and MT2 agonist, AMMTC, and blocked by the antagonist, CBPT. RORalpha receptors are also expressed in MCF-7 breast cancer cells and the putative RORalpha agonist CPG-52608 inhibits MCF-7 cell growth but with a very different dose-response than melatonin. Finally, melatonin and AMMTC, but not CPG-52608, can repress RORalpha transcriptional activity in MCF-7 cells.
Collapse
MESH Headings
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carbazoles/pharmacology
- Cell Division/drug effects
- Dose-Response Relationship, Drug
- Humans
- Immunoblotting
- Luciferases/drug effects
- Luciferases/genetics
- Luciferases/metabolism
- Melatonin/pharmacology
- Nuclear Receptor Subfamily 1, Group F, Member 1
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cell Surface/drug effects
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Cytoplasmic and Nuclear/drug effects
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Melatonin
- Recombinant Fusion Proteins/drug effects
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Thiazoles/pharmacology
- Thiosemicarbazones/pharmacology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- P T Ram
- Department of Structural and Cellular Biology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Nowfar S, Teplitzky SR, Melancon K, Kiefer TL, Cheng Q, Dwived PD, Bischoff ED, Moro K, Anderson MB, Dai J, Lai L, Yuan L, Hill SM. Tumor prevention by 9-cis-retinoic acid in the N-nitroso-N-methylurea model of mammary carcinogenesis is potentiated by the pineal hormone melatonin. Breast Cancer Res Treat 2002; 72:33-43. [PMID: 12004806 DOI: 10.1023/a:1014912919470] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Our laboratory has demonstrated that treatment of MCF-7 breast cancer cells with melatonin (Mlt) followed 24h later with physiological concentrations of all-trans retinoic acid (atRA) results in apoptosis. These studies were extended into trials using the N-nitroso-N-methylurea (NMU)-induced rat mammary tumor model. Initial studies conducted by feeding the animals 9-cis-retinoic acid (9cRA in the chow) and administering melatonin by subcutaneous injection in the late afternoon demonstrated that the combination of Mlt and 9cRA was able to significantly prevent tumor development, and that the combination was more efficacious that either Mlt or 9cRA alone. In this report, we conducted studies to determine if lower doses of 9cRA could be used in combination with Mlt while still maintaining anti-tumor activity and if the route of administration of 9cRA (bolus (gavage) v.s. chronic (chow) routes) affected its interaction with Mlt. The studies presented here demonstrate that significantly reduced doses of 9cRA can be used in combination with Mlt while maintaining anti-tumor efficacy. Furthermore, our studies demonstrate that 9cRA is equally effective when it is administered chronically (chow) or as a bolus (gavage). These data demonstrate that the combined use of Mlt and 9cRA produces additive or synergistic effects, which are more efficacious than 9cRA alone. This combination of Mlt and 9cRA could be a potentially useful clinical treatment regimen for breast cancer since it allows the use of lower doses of retinoic acid, thus, avoiding the toxic side effects associated with the use of high dose retinoids.
Collapse
Affiliation(s)
- S Nowfar
- Department of Structural & Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|