301
|
Park JH, Seo I, Shim HM, Cho H. Melatonin ameliorates SGLT2 inhibitor-induced diabetic ketoacidosis by inhibiting lipolysis and hepatic ketogenesis in type 2 diabetic mice. J Pineal Res 2020; 68:e12623. [PMID: 31743484 DOI: 10.1111/jpi.12623] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022]
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are effective hypoglycemic agents that can induce glycosuria. However, there are increasing concerns that they might induce diabetic ketoacidosis. This study investigated the effect of melatonin on SGTL2i-induced ketoacidosis in insulin-deficient type 2 diabetic (T2D) mice. The SGLT2i dapagliflozin reduced blood glucose level and plasma insulin concentrations in T2D mice, but induced increases in the concentrations of plasma β-hydroxybutyrate, acetoacetate, and free fatty acid and a decrease in the concentration of plasma bicarbonate, resulting in ketoacidosis. Melatonin inhibited dapagliflozin-induced ketoacidosis without inducing any change in blood glucose level or plasma insulin concentration. In white adipose tissue, melatonin inhibited lipolysis and downregulated phosphorylation of PKA, HSL, and perilipin-1. In liver tissue, melatonin suppressed cellular cyclic AMP levels and downregulated phosphorylation of PKA, AMPK, and acetyl-CoA carboxylase (ACC). In addition, melatonin increased hepatic ACC activity, but decreased hepatic CPT1a activity and acetyl-CoA content. These effects of melatonin on lipolysis and hepatic ketogenesis were blocked by pretreatment with melatonin receptor antagonist or PKA activator. Collectively, these results suggest that melatonin can ameliorate SGLT2i-induced ketoacidosis by inhibiting lipolysis and hepatic ketogenesis though cyclic AMP/PKA signaling pathways in T2D mice. Thus, melatonin treatment may offer protection against SGLT2i-induced ketoacidosis.
Collapse
Affiliation(s)
- Jae-Hyung Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Incheol Seo
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Hae-Min Shim
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Hochan Cho
- Division of Endocrinology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
302
|
Bolteau R, Descamps F, Ettaoussi M, Caignard DH, Delagrange P, Melnyk P, Yous S. Quinazoline and phthalazine derivatives as novel melatonin receptor ligands analogues of agomelatine. Eur J Med Chem 2020; 189:112078. [PMID: 32004937 DOI: 10.1016/j.ejmech.2020.112078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/12/2019] [Accepted: 01/16/2020] [Indexed: 01/02/2023]
Abstract
For further development of successors of Agomelatine through modulation of its pharmacokinetic properties, we report herein the design, synthesis and pharmacological results of a new family of melatonin receptor ligands. Issued from the introduction of quinazoline and phthalazine scaffolds carrying an ethyl amide lateral chain and a methoxy group as bioisosteric ligands analogues of previously developed Agomelatine. The biological activity of the prepared analogues was compared with that of Agomelatine. Quinazoline and phthalazine rings proved to be a versatile scaffold for easy feasible MT1 and MT2 ligands. Potent agonists with sub-micromolar binding affinity were obtained. However, the presence of two nitrogen atoms resulted in compounds with lower affinity for both MT1 and MT2, in comparison with the parent compound, balanced by the exhibition of good pharmacokinetic properties.
Collapse
Affiliation(s)
- Raphaël Bolteau
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000, Lille, France
| | - Florian Descamps
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000, Lille, France
| | - Mohamed Ettaoussi
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000, Lille, France.
| | - Daniel H Caignard
- PEX Biotechnologie Chimie & Biologie, Institut de Recherches Servier, 78290, Croissy sur Seine, France; Institut de Recherches Internationales Servier, 92150, Suresnes, France
| | - Philippe Delagrange
- PEX Biotechnologie Chimie & Biologie, Institut de Recherches Servier, 78290, Croissy sur Seine, France
| | - Patricia Melnyk
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000, Lille, France
| | - Saïd Yous
- Univ. Lille, Inserm, CHU Lille, UMR-S1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000, Lille, France.
| |
Collapse
|
303
|
Merlo S, Luaces JP, Spampinato SF, Toro-Urrego N, Caruso GI, D’Amico F, Capani F, Sortino MA. SIRT1 Mediates Melatonin's Effects on Microglial Activation in Hypoxia: In Vitro and In Vivo Evidence. Biomolecules 2020; 10:biom10030364. [PMID: 32120833 PMCID: PMC7175216 DOI: 10.3390/biom10030364] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/31/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023] Open
Abstract
Melatonin exerts direct neuroprotection against cerebral hypoxic damage, but the mechanisms of its action on microglia have been less characterized. Using both in vitro and in vivo models of hypoxia, we here focused on the role played by silent mating type information regulation 2 homolog 1 (SIRT1) in melatonin's effects on microglia. Viability of rat primary microglia or microglial BV2 cells and SH-SY5Y neurons was significantly reduced after chemical hypoxia with CoCl2 (250 μM for 24 h). Melatonin (1 μM) significantly attenuated CoCl2 toxicity on microglia, an effect prevented by selective SIRT1 inhibitor EX527 (5 μM) and AMP-activated protein kinase (AMPK) inhibitor BML-275 (2 μM). CoCl2 did not modify SIRT1 expression, but prevented nuclear localization, while melatonin appeared to restore it. CoCl2 induced nuclear localization of hypoxia-inducible factor-1α (HIF-1α) and nuclear factor-kappa B (NF-kB), an effect contrasted by melatonin in an EX527-dependent fashion. Treatment of microglia with melatonin attenuated potentiation of neurotoxicity. Common carotid occlusion was performed in p7 rats, followed by intraperitoneal injection of melatonin (10 mg/kg). After 24 h, the number of Iba1+ microglia in the hippocampus of hypoxic rats was significantly increased, an effect not prevented by melatonin. At this time, SIRT1 was only detectable in the amoeboid, Iba1+ microglial population selectively localized in the corpus callosum. In these cells, nuclear localization of SIRT1 was significantly lower in hypoxic animals, an effect prevented by melatonin. NF-kB showed an opposite expression pattern, where nuclear localization in Iba1+ cells was significantly higher in hypoxic, but not in melatonin-treated animals. Our findings provide new evidence for a direct effect of melatonin on hypoxic microglia through SIRT1, which appears as a potential pharmacological target against hypoxic-derived neuronal damage.
Collapse
Affiliation(s)
- Sara Merlo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy; (S.M.); (S.F.S.); (G.I.C.)
| | - Juan Pablo Luaces
- Laboratorio de Citoarquitectura y Plasticidad, Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1122, Argentina; (J.P.L.); (N.T.-U.); (F.C.)
| | - Simona Federica Spampinato
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy; (S.M.); (S.F.S.); (G.I.C.)
| | - Nicolas Toro-Urrego
- Laboratorio de Citoarquitectura y Plasticidad, Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1122, Argentina; (J.P.L.); (N.T.-U.); (F.C.)
| | - Grazia Ilaria Caruso
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy; (S.M.); (S.F.S.); (G.I.C.)
| | - Fabio D’Amico
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad, Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1122, Argentina; (J.P.L.); (N.T.-U.); (F.C.)
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy; (S.M.); (S.F.S.); (G.I.C.)
- Correspondence: ; Tel.: +39-095-4781192
| |
Collapse
|
304
|
Gu C, Yang H, Chang K, Zhang B, Xie F, Ye J, Chang R, Qiu X, Wang Y, Qu Y, Wang J, Li M. Melatonin alleviates progression of uterine endometrial cancer by suppressing estrogen/ubiquitin C/SDHB-mediated succinate accumulation. Cancer Lett 2020; 476:34-47. [PMID: 32061949 DOI: 10.1016/j.canlet.2020.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/06/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
Succinate is an important intermediate of the tricarboxylic acid cycle. Recently discovered roles of succinate demonstrate its involvement in immunity and cancer biology; however, the precise underlying mechanisms of its involvement in these additional roles remain to be determined. In the present study, succinate dehydrogenase (SDH) B was decreased in uterine endometrial cancer cells (UECC) under negative regulation of estrogen. This decrease was the result of lower expression levels of ubiquitin C (UBC), which was associated with the activation of peroxisome proliferator-activated receptor gamma and specificity protein 1. The decreased levels of SDHB resulted in the accumulation of succinate in UECC, and thus, a decrease in the production of fumaric acid. Succinate downregulated voltage-gated potassium channel subfamily Q member 1 (KCNQ1) levels by activating serum/glucocorticoid regulated kinase 1 and promoted the growth of UECC in vitro and in vivo. Treatment with melatonin restricted estrogen/UBC/SDHB-induced succinate accumulation and upregulated expression of KCNQ1 and reduced the succinate-mediated growth of UECC in vitro and in vivo. Furthermore, overexpression of melatonin receptor 1B amplified the inhibitory effects of melatonin on succinate-mediated UECC growth. Together, the data in the present study suggest that melatonin suppresses UECC progression by inhibiting estrogen/UBC/SDHB-induced succinate accumulation. The present study provides a scientific basis for potential therapeutic strategies and targets in UEC, particularly for patients with abnormally low levels of SDHB.
Collapse
Affiliation(s)
- Chunjie Gu
- NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China; Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China
| | - Huili Yang
- NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China; Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China
| | - Kaikai Chang
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China; Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, People's Republic of China
| | - Bing Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu Province, People's Republic of China
| | - Feng Xie
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China.
| | - Jiangfeng Ye
- Clinical Epidemiology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, People's Republic of China
| | - Ruiqi Chang
- NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China
| | - Xuemin Qiu
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China
| | - Yan Wang
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China
| | - Yuqing Qu
- Department of Pathology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China
| | - Jian Wang
- NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China
| | - Mingqing Li
- NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China; Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200080, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
305
|
Stein RM, Kang HJ, McCorvy JD, Glatfelter GC, Jones AJ, Che T, Slocum S, Huang XP, Savych O, Moroz YS, Stauch B, Johansson LC, Cherezov V, Kenakin T, Irwin JJ, Shoichet BK, Roth BL, Dubocovich ML. Virtual discovery of melatonin receptor ligands to modulate circadian rhythms. Nature 2020; 579:609-614. [PMID: 32040955 PMCID: PMC7134359 DOI: 10.1038/s41586-020-2027-0] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 01/31/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Reed M Stein
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Hye Jin Kang
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John D McCorvy
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Grant C Glatfelter
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), The State University of New York, Buffalo, NY, USA.,Designer Drug Research Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, USA
| | - Anthony J Jones
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), The State University of New York, Buffalo, NY, USA
| | - Tao Che
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Samuel Slocum
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xi-Ping Huang
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Yurii S Moroz
- National Taras Shevchenko University of Kyiv, Kiev, Ukraine.,Chemspace, Monmouth Junction, NJ, USA
| | - Benjamin Stauch
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA.,Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Linda C Johansson
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA.,Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Vadim Cherezov
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA.,Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Terry Kenakin
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John J Irwin
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
| | - Bryan L Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Margarita L Dubocovich
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo (SUNY), The State University of New York, Buffalo, NY, USA.
| |
Collapse
|
306
|
Melatonin as an Agent for Direct Pulp-Capping Treatment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17031043. [PMID: 32041360 PMCID: PMC7037898 DOI: 10.3390/ijerph17031043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/31/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022]
Abstract
Melatonin plays an essential role in the regulation of bone growth. The actions that melatonin exerts on odontoblasts may be similar to its action on osteoblasts. This research aimed to evaluate the pulp response to melatonin used for direct pulp capping to evaluate the antioxidant effect of melatonin administered orally and its influence on dental pulp. Direct pulp capping was performed on the upper molars of Sprague Dawley rats using melatonin or Mineral Trioxide Aggregate (MTA). The study groups were: MTA; Melatonin; MTA + Melatonin administered orally; and Melatonin + Melatonin administered orally. In the latter two groups, the animals drank water dosed with melatonin ad libitum (10 mg/100 mL). After 30 days, the animals were sacrificed, and 5 ml of blood, the kidneys, and the liver were extracted in order to evaluate oxidative stress using thiobarbituric acid reactive substances testing (TBARS). Fragments of the maxilla containing the study molars were prepared for histological evaluation. The degree of pulp inflammation and pulp necrosis, the presence of reparative dentin and dentin bridging the pulp chamber, the presence and regularity of the odontoblastic layer, and the presence of pulp fibrosis were evaluated. No significant differences were found between the four study groups for any of the studied histological variables. The oral administration of melatonin did not modify the local effects of MTA or melatonin on dental pulp, or reduce basal-level oxidative stress. The effect of melatonin on pulp is similar to that of MTA and may be used as an agent for direct pulp capping.
Collapse
|
307
|
Colunga Biancatelli RML, Berrill M, Mohammed YH, Marik PE. Melatonin for the treatment of sepsis: the scientific rationale. J Thorac Dis 2020; 12:S54-S65. [PMID: 32148926 DOI: 10.21037/jtd.2019.12.85] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sepsis affects 30 million people worldwide, leading to 6 million deaths every year (WHO), and despite decades of research, novel initiatives are drastically needed. According to the current literature, oxidative imbalance and mitochondrial dysfunction are common features of septic patients that can cause multiorgan failure and death. Melatonin, alongside its traditionally accepted role as the master hormonal regulator of the circadian rhythm, is a promising adjunctive drug for sepsis through its anti-inflammatory, antiapoptotic and powerful antioxidant properties. Several animal models of sepsis have demonstrated that melatonin can prevent multiorgan dysfunction and improve survival through restoring mitochondrial electron transport chain (ETC) function, inhibiting nitric oxide synthesis and reducing cytokine production. The purpose of this article is to review the current evidence for the role of melatonin in sepsis, review its pharmacokinetic profile and virtual absence of side effects. While clinical data is limited, we propose the adjunctive use of melatonin is patients with severe sepsis and septic shock.
Collapse
Affiliation(s)
- Ruben Manuel Luciano Colunga Biancatelli
- Division of Pulmonary and Critical Care Medicine, Eastern Virginia Medical School, Norfolk, VA, USA.,Policlinico Umberto I, La Sapienza University of Rome, Rome, Italy
| | - Max Berrill
- Division of Pulmonary and Critical Care Medicine, Eastern Virginia Medical School, Norfolk, VA, USA.,St. Peter's Hospital, Department of Respiratory Medicine, London, UK
| | - Yassen H Mohammed
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Paul E Marik
- Division of Pulmonary and Critical Care Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
308
|
Melatonin inhibits GABAergic neurons in the hypothalamus consistent with a reduction in wakefulness. Neuroreport 2020; 31:92-98. [DOI: 10.1097/wnr.0000000000001374] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
309
|
D'Occhio MJ, Ghuman SS, Neglia G, Della Valle G, Baruselli PS, Zicarelli L, Visintin JA, Sarkar M, Campanile G. Exogenous and endogenous factors in seasonality of reproduction in buffalo: A review. Theriogenology 2020; 150:186-192. [PMID: 32000994 DOI: 10.1016/j.theriogenology.2020.01.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/29/2022]
Abstract
Seasonal breeding in buffalo is influenced by exogenous (photoperiod, climate, nutrition, management) and endogenous (hormones, genotype) factors. Buffalo are negatively photoperiodic and show a natural increase in fertility during decreasing day length. The hormone melatonin is produced by the pineal gland and has a fundamental role in photoperiodic time measurement within the brain. This drives annual cycles of gonadotropin secretion and gonadal function in buffaloes. Some melatonin is released into the systemic circulation and, together with peripherally produced melatonin, acts at somatic tissues. In the ovaries and testes of buffalo, melatonin acts as an antioxidant and scavenges oxygen free radicals to reduce both oxidative stress and apoptosis. This has beneficial effects on gametogenesis and steroidogenesis. Female buffalo treated with melatonin show an improved response to estrus synchronization protocols in out-of-season breeding. Melatonin acts through melatonin receptors MT1 and MT2 and the gene for MT1 (MTNR1A) is polymorphic in buffaloes. Single nucleotide polymorphisms (SNPs) in gene MTNR1A have been associated with fertility in female buffalo. The knowledge and tools are available to lift the reproductive performance of buffalo. This is highly important as the global demand for nutritious buffalo food products has undergone a sharp rise, and continues to grow. Buffalo can make an important contribution to affordable, nutritious animal protein. This will help address global nutritional security.
Collapse
Affiliation(s)
- Michael J D'Occhio
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Sarvpreet S Ghuman
- Department of Teaching Veterinary Clinical Complex, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, 141004, India
| | - Gianluca Neglia
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy.
| | - Giovanni Della Valle
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Pietro S Baruselli
- Department of Animal Reproduction, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Luigi Zicarelli
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - José A Visintin
- Department of Animal Reproduction, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Mihir Sarkar
- Physiology and Climatology Division, Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, 243122, India
| | - Giuseppe Campanile
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| |
Collapse
|
310
|
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: 2.0] [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
|
311
|
Kalmukova OO, Yurchenko AV, Savchuk AM, Dzerzhynsky ME. Changes in the Inflammatory Status in White Adipose Tissue of Rats with Diet-Induced Obesity at Different Regimens of Melatonin Administration. CYTOL GENET+ 2020. [DOI: 10.3103/s0095452720010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
312
|
Wang SY, Shi XC, Laborda P. Indole-based melatonin analogues: Synthetic approaches and biological activity. Eur J Med Chem 2020; 185:111847. [DOI: 10.1016/j.ejmech.2019.111847] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022]
|
313
|
Bizzarri M. Advances in Characterizing Recently-Identified Molecular Actions of Melatonin: Clinical Implications. APPROACHING COMPLEX DISEASES 2020. [PMCID: PMC7164543 DOI: 10.1007/978-3-030-32857-3_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Melatonin, N-acetyl-5-methoxy-tryptamine, was discovered to be a product of serotonin metabolism in the mammalian pineal gland where its synthesis is under control of the light:dark cycle. Besides its regulatory pathway involving ganglion cells in the retina, the neural connections between the eyes and the pineal gland include the master circadian clock, the suprachiasmatic nuclei, and the central and peripheral nervous systems. Since pineal melatonin is released into the blood and into the cerebrospinal fluid, it has access to every cell in an organism and it mediates system-wide effects. Subsequently, melatonin was found in several extrapineal organs and, more recently, perhaps in every cell of every organ. In contrast to the pinealocytes, non-pineal cells do not discharge melatonin into the blood; rather it is used locally in an intracrine, autocrine, or paracrine manner. Melatonin levels in non-pineal cells do not exhibit a circadian rhythm and do not depend on circulating melatonin concentrations although when animals are treated with exogenous melatonin it is taken up by presumably all cells. Mitochondria are the presumed site of melatonin synthesis in all cells; the enzymatic machinery for melatonin synthesis has been identified in mitochondria. The association of melatonin with mitochondria, because of its ability to inhibit oxidative stress, is very fortuitous since these organelles are a major site of damaging reactive oxygen species generation. In this review, some of the actions of non-pineal-derived melatonin are discussed in terms of cellular and subcellular physiology.
Collapse
Affiliation(s)
- Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
314
|
Ciccone CD. Geriatric Pharmacology. GUCCIONE'S GERIATRIC PHYSICAL THERAPY 2020:102-136. [DOI: 10.1016/b978-0-323-60912-8.00006-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
315
|
Bjørklund G, Rajib SA, Saffoon N, Pen JJ, Chirumbolo S. Insights on Melatonin as an Active Pharmacological Molecule in Cancer Prevention: What's New? Curr Med Chem 2019; 26:6304-6320. [PMID: 29714136 DOI: 10.2174/0929867325666180501094850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/10/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022]
Abstract
Along with playing an important role in circadian rhythm, melatonin is thought to play a significant role in preventing cells from damage, as well as in the inhibition of growth and in triggering apoptosis in malignant cells. Its relationship with circadian rhythms, energetic homeostasis, diet, and metabolism, is fundamental to achieve a better comprehension of how melatonin has been considered a chemopreventive molecule, though very few papers dealing with this issue. In this article, we tried to review the most recent evidence regarding the protective as well as the antitumoral mechanisms of melatonin, as related to diet and metabolic balance. From different studies, it was evident that an intracellular antioxidant defense mechanism is activated by upregulating an antioxidant gene battery in the presence of high-dose melatonin in malignant cells. Like other broad-spectrum antioxidant molecules, melatonin plays a vital role in killing tumor cells, preventing metastasis, and simultaneously keeping normal cells protected from oxidative stress and other types of tissue damage.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| | | | - Nadia Saffoon
- Department of Pharmacy and Forensic Science, Faculty of Life Science and Medicine, King's College London, London, United Kingdom
| | - Joeri J Pen
- Diabetes Clinic, Department of Internal Medicine, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Department of Nutrition, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
316
|
Barbosa-Méndez S, Salazar-Juárez A. Melatonin decreases cocaine-induced locomotor activity in pinealectomized rats. ACTA ACUST UNITED AC 2019; 42:295-308. [PMID: 31859790 PMCID: PMC7236171 DOI: 10.1590/1516-4446-2018-0400] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 09/13/2019] [Indexed: 12/22/2022]
Abstract
Objective: Several studies have shown that the time of day regulates the reinforcing effects of cocaine. Additionally, melatonin and its MT1 and MT2 receptors have been found to participate in modulation of the reinforcing effects of such addictive drugs as cocaine. Loss of the diurnal variation in cocaine-induced locomotor sensitization and cocaine-induced place preference has been identified in pinealectomized mice. In addition, several studies in rodents have shown that administration of melatonin decreased the reinforcing effects of cocaine. The objective of this study was to evaluate the effect of melatonin on cocaine-induced locomotor activity in pinealectomized rats at different times of day (zeitgeber time [ZT]4, ZT10, ZT16, and ZT22). Methods: Naïve, pinealectomized Wistar rats received cocaine at different times of day. Melatonin was administered 30 min before cocaine; luzindole was administered 15 min prior to melatonin and 45 min before cocaine. After administration of each treatment, locomotor activity for each animal was recorded for a total of 30 min. Pinealectomy was confirmed at the end of the experiment through melatonin quantitation by ELISA. Results: Cocaine-induced locomotor activity varied according to the time of day. Continuous lighting and pinealectomy increased cocaine-induced locomotor activity. Melatonin administration decreased cocaine-induced locomotor activity in naïve and pinealectomized rats at different times of day. Luzindole blocked the melatonin-induced reduction in cocaine-induced locomotor activity in pinealectomized rats. Conclusion: Given its ability to mitigate various reinforcing effects of cocaine, melatonin could be a useful therapy for cocaine abuse.
Collapse
Affiliation(s)
- Susana Barbosa-Méndez
- Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría, Ciudad de México, Mexico
| | - Alberto Salazar-Juárez
- Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría, Ciudad de México, Mexico
| |
Collapse
|
317
|
Barbosa-Méndez S, Salazar-Juarez A. Melatonin does not produce sedation in rats: A chronobiological study. Chronobiol Int 2019; 37:353-374. [DOI: 10.1080/07420528.2019.1702554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Susana Barbosa-Méndez
- Molecular Neurobiology and Neurochemistry of Addiction, Ramón de la Fuente Muñiz National Institute of Psychiatry, Ciudad de México, México
| | - Alberto Salazar-Juarez
- Molecular Neurobiology and Neurochemistry of Addiction, Ramón de la Fuente Muñiz National Institute of Psychiatry, Ciudad de México, México
| |
Collapse
|
318
|
Beker MC, Caglayan B, Caglayan AB, Kelestemur T, Yalcin E, Caglayan A, Kilic U, Baykal AT, Reiter RJ, Kilic E. Interaction of melatonin and Bmal1 in the regulation of PI3K/AKT pathway components and cellular survival. Sci Rep 2019; 9:19082. [PMID: 31836786 PMCID: PMC6910929 DOI: 10.1038/s41598-019-55663-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/22/2019] [Indexed: 01/08/2023] Open
Abstract
The circadian rhythm is driven by a master clock within the suprachiasmatic nucleus which regulates the rhythmic secretion of melatonin. Bmal1 coordinates the rhythmic expression of transcriptome and regulates biological activities, involved in cell metabolism and aging. However, the role of Bmal1 in cellular- survival, signaling, its interaction with intracellular proteins, and how melatonin regulates its expression is largely unclear. Here we observed that melatonin increases the expression of Bmal1 and both melatonin and Bmal1 increase cellular survival after oxygen glucose deprivation (OGD) while the inhibition of Bmal1 resulted in the decreased cellular survival without affecting neuroprotective effects of melatonin. By using a planar surface immunoassay for PI3K/AKT signaling pathway components, we revealed that both melatonin and Bmal1 increased phosphorylation of AKT, ERK-1/2, PDK1, mTOR, PTEN, GSK-3αβ, and p70S6K. In contrast, inhibition of Bmal1 resulted in decreased phosphorylation of these proteins, which the effect of melatonin on these signaling molecules was not affected by the absence of Bmal1. Besides, the inhibition of PI3K/AKT decreased Bmal1 expression and the effect of melatonin on Bmal1 after both OGD in vitro and focal cerebral ischemia in vivo. Our data demonstrate that melatonin controls the expression of Bmal1 via PI3K/AKT signaling, and Bmal1 plays critical roles in cellular survival via activation of survival kinases.
Collapse
Affiliation(s)
- Mustafa C Beker
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Berrak Caglayan
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Medical Biology, International School of Medicine, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Ahmet B Caglayan
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Taha Kelestemur
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Esra Yalcin
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Aysun Caglayan
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, 34810, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Ulkan Kilic
- Department of Medical Biology, School of Medicine, University of Health Sciences, 34668, Istanbul, Turkey
| | - Ahmet T Baykal
- Department of Medical Biochemistry, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752, Istanbul, Turkey
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, 78229, Texas, USA
| | - Ertugrul Kilic
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, 34810, Istanbul, Turkey.
- Department of Physiology, School of Medicine, Istanbul Medipol University, 34810, Istanbul, Turkey.
| |
Collapse
|
319
|
Cecon E, Liu L, Jockers R. Melatonin receptor structures shed new light on melatonin research. J Pineal Res 2019; 67:e12606. [PMID: 31442321 DOI: 10.1111/jpi.12606] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/17/2019] [Accepted: 08/18/2019] [Indexed: 01/02/2023]
Abstract
The tryptophan derivative melatonin is an evolutionary old molecule that is involved in a pleiotropy of physiological functions. In humans, age-related decline of circulating melatonin levels and/or dysregulation of its circadian synthesis pattern have been associated with several disorders and disease states. Several molecular targets have been proposed for melatonin since its discovery, in 1959. Among them, melatonin MT1 and MT2 receptors are the best characterized melatonin targets, mediating melatonin effects in a variety of tissues. They belong to the superfamily of G protein-coupled receptors. Two back-to-back articles published in the "Nature" Journal earlier this year present the first crystal structures of the human MT1 and MT2 in its inactive states. Here, we will briefly outline the discovery path of melatonin receptors until their structural elucidation and discuss how these new findings will guide future research toward a better understanding of their function and rational drug design.
Collapse
MESH Headings
- Animals
- History, 20th Century
- History, 21st Century
- Humans
- Melatonin/chemistry
- Melatonin/history
- Melatonin/metabolism
- Protein Structure, Tertiary
- Receptor, Melatonin, MT1/chemistry
- Receptor, Melatonin, MT1/metabolism
- Receptor, Melatonin, MT2/chemistry
- Receptor, Melatonin, MT2/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Erika Cecon
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Lei Liu
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Ralf Jockers
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| |
Collapse
|
320
|
Li DJ, Tong J, Li YH, Meng HB, Ji QX, Zhang GY, Zhu JH, Zhang WJ, Zeng FY, Huang G, Hua X, Shen FM, Wang P. Melatonin safeguards against fatty liver by antagonizing TRAFs-mediated ASK1 deubiquitination and stabilization in a β-arrestin-1 dependent manner. J Pineal Res 2019; 67:e12611. [PMID: 31541591 DOI: 10.1111/jpi.12611] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/17/2019] [Accepted: 09/14/2019] [Indexed: 12/15/2022]
Abstract
Melatonin has been previously shown to prevent nonalcoholic fatty liver disease (NAFLD), yet the underlying mechanisms are poorly understood. Here, we identified a previously unknown regulatory action of melatonin on apoptosis signal-regulating kinase 1 (ASK1) signaling pathway in the pathogenesis and development of NAFLD. Although melatonin administration did not alter food intake, it significantly alleviated fatty liver phenotypes, including the body weight gain, insulin resistance, hepatic lipid accumulation, steatohepatitis, and fibrosis in a high-fat diet (HFD)-induced NAFLD mouse model (in vivo). The protection of melatonin against NAFLD was not affected by inactivation of Kupffer cell in this model. In NAFLD mice liver, ASK1 signal cascade was substantially activated, evidence by the enhancement of total ASK1, phospho-ASK1, phospho-MKK3/6, phospho-p38, phospho-MKK4/7, and phospho-JNK. Melatonin treatment significantly suppressed the ASK1 upregulation and the phosphorylation of ASK1, MKK3/6, MKK4/7, p38, and JNK. Mechanistically, we found that lipid stress triggered the interaction between ASK1 and TNF receptor-associated factors (TRAFs), including TRAF1, TRAF2, and TRAF6, which resulted in ASK1 deubiquitination and thereby increased ASK1 protein stability. Melatonin did not alter ASK1 mRNA level; however, it activated a scaffold protein β-arrestin-1 and enabled it to bind to ASK1, which antagonized the TRAFs-mediated ASK1 deubiquitination, and thus reduced ASK1 protein stability. Consistent with these findings, knockout of β-arrestin-1 in mice partly abolished the protection of melatonin against NAFLD. Taken together, our results for the first time demonstrate that melatonin safeguards against NAFLD by eliminating ASK1 activation via inhibiting TRAFs-mediated ASK1 deubiquitination and stabilization in a β-arrestin-1 dependent manner.
Collapse
Affiliation(s)
- Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Yong-Hua Li
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hong-Bo Meng
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing-Xin Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Guo-Yan Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Jia-Hui Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Wen-Jing Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Fei-Yan Zeng
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Gang Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xia Hua
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Pei Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Tongji University School of Medicine, Tongji University, Shanghai, China
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
321
|
The Neuroprotective Effects of Melatonin: Possible Role in the Pathophysiology of Neuropsychiatric Disease. Brain Sci 2019; 9:brainsci9100285. [PMID: 31640239 PMCID: PMC6826722 DOI: 10.3390/brainsci9100285] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 11/17/2022] Open
Abstract
Melatonin is a hormone that is secreted by the pineal gland. To date, melatonin is known to regulate the sleep cycle by controlling the circadian rhythm. However, recent advances in neuroscience and molecular biology have led to the discovery of new actions and effects of melatonin. In recent studies, melatonin was shown to have antioxidant activity and, possibly, to affect the development of Alzheimer's disease (AD). In addition, melatonin has neuroprotective effects and affects neuroplasticity, thus indicating potential antidepressant properties. In the present review, the new functions of melatonin are summarized and a therapeutic target for the development of new drugs based on the mechanism of action of melatonin is proposed.
Collapse
|
322
|
Dampening of neurotransmitter action: molecular similarity within the melatonin structure. Endocr Regul 2019; 52:199-207. [PMID: 31517615 DOI: 10.2478/enr-2018-0025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVES Melatonin initiates physiologic and therapeutic responses in various tissues through binding to poorly defined MT receptors regulated by G-proteins and purine nucleotides. Melatonin's interaction with other G-protein regulated receptors, including those of serotonin, is unclear. This study explores the potential for the interaction of melatonin with nucleotide and receptor ligand structures. METHODS The study uses a computational program to investigate relative molecular similarity by the comparative superimposition and quantitative fitting of molecular structures to adenine and guanine nucleotide templates. RESULTS A minimum energy melatonin conformer replicates the nucleotide fits of ligand structures that regulate Gαi and Gαq proteins via serotonin, dopamine, opioid, α-adrenoceptor, and muscarinic receptor classes. The same conformer also replicates the nucleotide fits of ligand structures regulating K+ and Ca2+ ion channels. The acyl-methoxy distance within the melatonin conformer matches a carbonyl-hydroxyl distance in guanine nucleotide. CONCLUSION Molecular similarity within the melatonin and ligand structures relates to the established effects of melatonin on cell receptors regulated by purine nucleotides in cell signal transduction processes. Pharmacologic receptor promiscuity may contribute to the widespread effects of melatonin.
Collapse
|
323
|
|
324
|
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: 2.5] [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
|
325
|
Han D, Wang Y, Chen J, Zhang J, Yu P, Zhang R, Li S, Tao B, Wang Y, Qiu Y, Xu M, Gao E, Cao F. Activation of melatonin receptor 2 but not melatonin receptor 1 mediates melatonin-conferred cardioprotection against myocardial ischemia/reperfusion injury. J Pineal Res 2019; 67:e12571. [PMID: 30903623 DOI: 10.1111/jpi.12571] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/01/2019] [Accepted: 03/17/2019] [Indexed: 12/14/2022]
Abstract
Accumulated pieces of evidence have proved the beneficial effects of melatonin on myocardial ischemia/reperfusion (MI/R) injury, and these effects were largely dependent on melatonin membrane receptor activation. In humans and other mammals, there are two types of melatonin receptors, including the melatonin receptor 1 (MT1, melatonin receptor 1a or MTNR1A) and melatonin receptor 1 (MT2, melatonin receptor 1b or MTNR1B) receptor subtypes. However, which receptor mediates melatonin-conferred cardioprotection remains unclear. In this study, we employed both loss-of-function and gain-of-function approaches to reveal the answer. Mice (wild-type; MT1 or MT2 silencing by in vivo minicircle vector; and those overexpressing MT1 or MT2 by in vivo AAV9 vector) were exposed to MI/R injury. Both MT1 and MT2 were present in wild-type myocardium. MT2, but not MT1, was essentially upregulated after MI/R Melatonin administration significantly reduced myocardial injury and improved cardiac function after MI/R Mechanistically, melatonin treatment suppressed MI/R-initiated myocardial oxidative stress and nitrative stress, alleviated endoplasmic reticulum stress and mitochondrial injury, and inhibited myocardial apoptosis. These beneficial actions of melatonin were absent in MT2-silenced heart, but not the MT1 subtype. Furthermore, AAV9-mediated cardiomyocyte-specific overexpression of MT2, but not MT1, mitigated MI/R injury and improved cardiac dysfunction, which was accompanied by significant amelioration of oxidative stress, endoplasmic reticulum stress, and mitochondrial dysfunction. Mechanistically, MT2 protected primary cardiomyocytes against hypoxia/reoxygenation injury via MT2/Notch1/Hes1/RORα signaling. Our study presents the first direct evidence that the MT2 subtype, but not MT1, is a novel endogenous cardiac protective receptor against MI/R injury. Medications specifically targeting MT2 may hold promise in fighting ischemic heart disease.
Collapse
MESH Headings
- Animals
- Apoptosis
- Disease Models, Animal
- Endoplasmic Reticulum Stress/genetics
- Humans
- Male
- Mice
- Myocardial Reperfusion Injury/genetics
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Oxidative Stress/genetics
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT1/metabolism
- Receptor, Melatonin, MT2/genetics
- Receptor, Melatonin, MT2/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Dong Han
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yongjun Wang
- Department of Cardiovascular Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangwei Chen
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jibin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng Yu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ran Zhang
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuang Li
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, China
| | - Bo Tao
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yabin Wang
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ya Qiu
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mengqi Xu
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Feng Cao
- Department of Cardiology, National Clinical Research Center for Geriatric Diseases, 2nd Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
326
|
Zhao CN, Wang P, Mao YM, Dan YL, Wu Q, Li XM, Wang DG, Davis C, Hu W, Pan HF. Potential role of melatonin in autoimmune diseases. Cytokine Growth Factor Rev 2019; 48:1-10. [DOI: 10.1016/j.cytogfr.2019.07.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/28/2022]
|
327
|
Klosen P, Lapmanee S, Schuster C, Guardiola B, Hicks D, Pevet P, Felder-Schmittbuhl MP. MT1 and MT2 melatonin receptors are expressed in nonoverlapping neuronal populations. J Pineal Res 2019; 67:e12575. [PMID: 30937953 DOI: 10.1111/jpi.12575] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/07/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022]
Abstract
Melatonin (MLT) exerts its physiological effects principally through two high-affinity membrane receptors MT1 and MT2. Understanding the exact mechanism of MLT action necessitates the use of highly selective agonists/antagonists to stimulate/inhibit a given MLT receptor. The respective distribution of MT1 and MT2 within the CNS and elsewhere is controversial, and here we used a "knock-in" strategy replacing MT1 or MT2 coding sequences with a LacZ reporter. The data show striking differences in the distribution of MT1 and MT2 receptors in the mouse brain: whereas the MT1 subtype was expressed in very few structures (notably including the suprachiasmatic nucleus and pars tuberalis), MT2 subtype receptors were identified within numerous brain regions including the olfactory bulb, forebrain, hippocampus, amygdala and superior colliculus. Co-expression of the two subtypes was observed in very few structures, and even within these areas they were rarely present in the same individual cell. In conclusion, the expression and distribution of MT2 receptors are much more widespread than previously thought, and there is virtually no correspondence between MT1 and MT2 cellular expression. The precise phenotyping of cells/neurons containing MT1 or MT2 receptor subtypes opens new perspectives for the characterization of links between MLT brain targets, MLT actions and specific MLT receptor subtypes.
Collapse
Affiliation(s)
- Paul Klosen
- Institute for Cellular and Integrative Neurosciences (UPR 3212), CNRS and University of Strasbourg, Strasbourg, France
| | - Sarawut Lapmanee
- Institute for Cellular and Integrative Neurosciences (UPR 3212), CNRS and University of Strasbourg, Strasbourg, France
| | | | | | - David Hicks
- Institute for Cellular and Integrative Neurosciences (UPR 3212), CNRS and University of Strasbourg, Strasbourg, France
| | - Paul Pevet
- Institute for Cellular and Integrative Neurosciences (UPR 3212), CNRS and University of Strasbourg, Strasbourg, France
| | | |
Collapse
|
328
|
Nasiri E, Alizadeh A, Roushandeh AM, Gazor R, Hashemi-Firouzi N, Golipoor Z. Melatonin-pretreated adipose-derived mesenchymal stem cells efficeintly improved learning, memory, and cognition in an animal model of Alzheimer's disease. Metab Brain Dis 2019; 34:1131-1143. [PMID: 31129766 DOI: 10.1007/s11011-019-00421-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/17/2019] [Indexed: 12/19/2022]
Abstract
Currently, mesenchymal stem cells (MSCs) based therapy has extensive attraction for Alzheimer's disease (AD). However, low survival rate of MSCs after transplantation is a huge challenging. The current study aimed to improve adipose-derived MSCs (AD-MSCs)-based therapy by their pre-treatment with melatonin (MT) 'a well-known antioxidant' in an animal model of AD. In this study, after isolating rat AD-MSCs from the epididymal white adipose tissues, the cells were pretreated with 5μM of MT for 24 hours. Forty male Wistar rats were randomly allocated to control, sham, amyloid-beta (Aβ) peptide, AD-MSCs and MT-pretreated ADMSCs groups. The novel object recognition, passive avoidance test, Morris water maze and open field test were performed two months following the cell transplantation. The rats were sacrificed 69 days following cell therapy. The brain tissues were removed for histopathological analysis and also immunohistochemistry was performed for two Aβ1-42 and Iba1 proteins. It has been revealed that both AD-MSCs and MT-AD-MSCs migrated to brain tissues after intravenous transplantation. However, MT-ADMSCs significantly improved learning, memory and cognition compared with AD-MSCs (P<0.05). Furthermore, clearance of Aβ deposition and reduction of microglial cells were significantly increased in the MT-ADMSCs compared with AD-MSCs. Although stem cell therapy has been introduced as a promising strategy in neurodegenerative diseases, however, its therapeutic properties are limited. It is suggested that pretreatment of MSCs with melatonin partly would increase the cells efficiency and consequently could decrease AD complication including memory and cognition.
Collapse
Affiliation(s)
- Ebrahim Nasiri
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Akram Alizadeh
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Rouhollah Gazor
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Zoleikha Golipoor
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
329
|
Prognostic Impact of Melatonin Receptors MT1 and MT2 in Non-Small Cell Lung Cancer (NSCLC). Cancers (Basel) 2019; 11:cancers11071001. [PMID: 31319607 PMCID: PMC6679108 DOI: 10.3390/cancers11071001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Several studies have investigated the inhibitory effect of melatonin on lung cancer cells. There are no data available on the prognostic impact of melatonin receptors MT1 and MT2 in non-small cell lung cancer (NSCLC). Materials and Methods: Immunohistochemical studies of MT1 and MT2 were conducted on NSCLC (N = 786) and non-malignant lung tissue (NMLT) (N = 120) using tissue microarrays. Molecular studies were performed on frozen fragments of NSCLC (N = 62; real time PCR), NMLT (N = 24) and lung cancer cell lines NCI-H1703, A549 and IMR-90 (real time PCR, western blot). Results: The expression of both receptors was higher in NSCLC than in NMLT. Higher MT1 and MT2 expression levels (at protein and mRNA) were noted in squamous cell carcinomas (SCC) compared to adenocarcinomas (AC). MT1 immunoexpression decreased as both the tumour size and the cancer stage increased in the whole cohort, while MT2 decreased as the cancer stage increased, with lymph node involvement (in the whole study group) and increasing malignancy grade (in SCC). Higher expression of MT2 was associated with a favorable prognosis. MT2 was an independent prognostic factor for overall survival (OS) in all analyzed NSCLC and in smoking patients. Conclusions: Our observations may point to the potential prognostic significance of MT2 in NSCLC.
Collapse
|
330
|
Uchimura N, Nakatome K, Miyata K, Uchiyama M. Effect of ramelteon coadministered with antidepressant in patients with insomnia and major depressive disorder: an exploratory study. Sleep Biol Rhythms 2019. [DOI: 10.1007/s41105-019-00230-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
331
|
Abstract
For many years now a treatment mitigating the debilitating effects of jet lag has been sought. Rapid travel across time zones leads, in most people, to temporary symptoms, in particular poor sleep, daytime alertness and poor performance. Mis-timed circadian rhythms are considered to be the main factor underlying jet-lag symptoms, together with the sleep deprivation from long haul flights. Virtually all aspects of physiology are rhythmic, from cells to systems, and circadian rhythms are coordinated by a central pacemaker or clock in the suprachiasmatic nucleus (SCN) of the hypothalamus. The SCN adapts slowly to changes in time zone, and peripheral clocks or oscillators adapt at different rates, such that the organism is in a state of desynchrony from the external environment and internally. Light exposure is the main factor controlling the circadian system and needs to be considered together with any pharmacological interventions. This review covers the relatively new chronobiotic drugs, which can hasten adaptation of the circadian system, together with drugs directly affecting alertness and sleep propensity. No current treatment can instantly shift circadian phase to a new time zone; however, adaptation can be hastened. The melatoninergic drugs are promising but larger trials in real-life situations are needed. For short stopovers it is recommended to preserve sleep and alertness without necessarily modifying the circadian system. New research suggests that modification of clock function via genetic manipulation may one day have clinical applications.
Collapse
Affiliation(s)
- Josephine Arendt
- Faculty of Health and Medical Sciences (FHMS), University of Surrey, Guildford, Surrey, GU2 7XH, UK.
| |
Collapse
|
332
|
Martínez PN, Menéndez ST, Villaronga MDLÁ, Ubelaker DH, García-Pedrero JM, C Zapico S. "The big sleep: Elucidating the sequence of events in the first hours of death to determine the postmortem interval". Sci Justice 2019; 59:418-424. [PMID: 31256813 DOI: 10.1016/j.scijus.2019.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/24/2019] [Accepted: 03/03/2019] [Indexed: 12/20/2022]
Abstract
Recent developments on postmortem interval estimation (PMI) take an advantage of the autolysis process, pointing out to the analysis of the expression of apoptosis and autophagy genes towards this purpose. Oxidative stress plays a role in this signaling as a regulatory mechanism and/or as a consequence of cell death. Additionally, melatonin has been implicated on apoptosis and autophagy signaling, making melatonin a suitable target for PMI determination. The aim of this study was to investigate the early PMI through the analysis of the expression of autophagy genes as well as oxidative stress and melatonin receptor. Our results demonstrated a rapidly increased on the expression of autophagy genes according to the expected sequence of events, then a marked decrease in this expression, matched with the switch to the apoptosis signaling. These results revealed potential candidates to analyze the PMI in the first hours of death, helping to estimate the time-since-death.
Collapse
Affiliation(s)
- Paula Núñez Martínez
- Departamento de Biología Funcional (Área de Fisiología), Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Spain
| | - Sofía T Menéndez
- Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | - María de Los Ángeles Villaronga
- Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | - Douglas H Ubelaker
- Anthropology Department, NMNH, Smithsonian Institution, MRC 112, Washington, DC, USA
| | - Juana M García-Pedrero
- Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain; CIBER de Cáncer (CIBERONC), Madrid, Spain
| | - Sara C Zapico
- Anthropology Department, NMNH, Smithsonian Institution, MRC 112, Washington, DC, USA; Department of Chemistry and Biochemistry, International Forensic Research Institute, Florida International University, Miami, FL, USA.
| |
Collapse
|
333
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Potes Y, Shabeeb D, Musa AE. Modulation of apoptosis by melatonin for improving cancer treatment efficiency: An updated review. Life Sci 2019; 228:228-241. [DOI: 10.1016/j.lfs.2019.05.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022]
|
334
|
Pliss MG, Kuzmenko NV, Rubanova NS, Tsyrlin VA. Dose-Dependent Mechanisms of Melatonin on the Functioning of the Cardiovascular System and on the Behavior of Normotensive Rats of Different Ages. ADVANCES IN GERONTOLOGY 2019. [DOI: 10.1134/s2079057019030111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
335
|
Effect of ramelteon on insomnia severity: evaluation of patient characteristics affecting treatment response. Sleep Biol Rhythms 2019. [DOI: 10.1007/s41105-019-00224-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
336
|
|
337
|
XFEL structures of the human MT 2 melatonin receptor reveal the basis of subtype selectivity. Nature 2019; 569:289-292. [PMID: 31019305 PMCID: PMC6589158 DOI: 10.1038/s41586-019-1144-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 03/26/2019] [Indexed: 11/28/2022]
Abstract
The human MT11 and MT22 melatonin receptors are G protein-coupled receptors
(GPCRs) involved in the regulation of circadian rhythm and sleep
patterns3. Drug
development efforts target both receptors for treatment of insomnia, circadian
rhythm and mood disorders, and cancer3, while MT2 has also been implicated in type 2
diabetes (T2D)4,5. Here we report the X-ray Free Electron
Laser (XFEL) structures of the human MT2 receptor in complex with
agonists 2-phenylmelatonin (2-pmt) and ramelteon6 at resolutions of 2.8 Å and 3.3
Å, respectively, along with two structures of function-related mutants,
H2085.46A (superscripts represent the Ballesteros-Weinstein
residue numbering nomenclature7) and N862.50D, obtained in complex with 2-pmt.
Comparison of the MT2 structures with MT18
reveals that, despite the fact that the orthosteric ligand-binding site residues
are conserved, there are notable conformational variations as well as
differences in [3H]-melatonin dissociation kinetics that provide new
insights into the selectivity between melatonin receptor subtypes. In addition
to the membrane-buried lateral ligand entry channel that is also observed in
MT1, the MT2 structures reveal a narrow opening
towards the solvent in the extracellular part of the receptor. We provide
functional and kinetic data supporting a prominent role for the intramembrane
ligand entry in both receptors, while simultaneously suggesting the possibility
of an extracellular entry path in MT2. Our findings contribute to a
molecular understanding of melatonin receptor subtype selectivity and ligand
access modes, which are essential for the design of highly selective melatonin
tool compounds and therapeutic agents.
Collapse
|
338
|
Ferri F, Olivieri F, Cannataro R, Caroleo MC, Cione E. Phytomelatonin Regulates Keratinocytes Homeostasis Counteracting Aging Process. COSMETICS 2019; 6:27. [DOI: 10.3390/cosmetics6020027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Phytomelatonin (PM) gained the greatest interest for its application in agriculture and its use to improve human health conditions. PM based supplement has been shown to possess antioxidant capabilities because it functions as a free radical scavenger. Reactive Oxygen Species (ROS), induced by both intrinsic (peroxide production) and extrinsic (UV-radiation) factors are biochemical mediators crucial in skin aging. Skin aging is also regulated by specific microRNAs (miRs). Herein we have shown the effect of PM free radical scavengers on the human keratinocyte cell line HaCat and on ROS formation induced by both extrinsic and intrinsic factors as well as their capability to positively modulate a member of the hsa-miR-29 family linked to aging. Our result highlights the regulatory role of PM for the keratinocytes homeostasis.
Collapse
Affiliation(s)
| | | | | | - Maria Cristina Caroleo
- GalaScreen Srl University of Calabria-Italy, 87036 Calabria, Italy
- Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Rende (CS), 87036 Rende, Italy
| | - Erika Cione
- GalaScreen Srl University of Calabria-Italy, 87036 Calabria, Italy
- Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, Rende (CS), 87036 Rende, Italy
| |
Collapse
|
339
|
Zhao D, Yu Y, Shen Y, Liu Q, Zhao Z, Sharma R, Reiter RJ. Melatonin Synthesis and Function: Evolutionary History in Animals and Plants. Front Endocrinol (Lausanne) 2019; 10:249. [PMID: 31057485 PMCID: PMC6481276 DOI: 10.3389/fendo.2019.00249] [Citation(s) in RCA: 353] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/29/2019] [Indexed: 12/12/2022] Open
Abstract
Melatonin is an ancient molecule that can be traced back to the origin of life. Melatonin's initial function was likely that as a free radical scavenger. Melatonin presumably evolved in bacteria; it has been measured in both α-proteobacteria and in photosynthetic cyanobacteria. In early evolution, bacteria were phagocytosed by primitive eukaryotes for their nutrient value. According to the endosymbiotic theory, the ingested bacteria eventually developed a symbiotic association with their host eukaryotes. The ingested α-proteobacteria evolved into mitochondria while cyanobacteria became chloroplasts and both organelles retained their ability to produce melatonin. Since these organelles have persisted to the present day, all species that ever existed or currently exist may have or may continue to synthesize melatonin in their mitochondria (animals and plants) and chloroplasts (plants) where it functions as an antioxidant. Melatonin's other functions, including its multiple receptors, developed later in evolution. In present day animals, via receptor-mediated means, melatonin functions in the regulation of sleep, modulation of circadian rhythms, enhancement of immunity, as a multifunctional oncostatic agent, etc., while retaining its ability to reduce oxidative stress by processes that are, in part, receptor-independent. In plants, melatonin continues to function in reducing oxidative stress as well as in promoting seed germination and growth, improving stress resistance, stimulating the immune system and modulating circadian rhythms; a single melatonin receptor has been identified in land plants where it controls stomatal closure on leaves. The melatonin synthetic pathway varies somewhat between plants and animals. The amino acid, tryptophan, is the necessary precursor of melatonin in all taxa. In animals, tryptophan is initially hydroxylated to 5-hydroxytryptophan which is then decarboxylated with the formation of serotonin. Serotonin is either acetylated to N-acetylserotonin or it is methylated to form 5-methoxytryptamine; these products are either methylated or acetylated, respectively, to produce melatonin. In plants, tryptophan is first decarboxylated to tryptamine which is then hydroxylated to form serotonin.
Collapse
Affiliation(s)
- Dake Zhao
- Biocontrol Engineering Research Center of Plant Disease and Pest, Yunnan University, Kunming, China
- Biocontrol Engineering Research Center of Crop Disease and Pest, Yunnan University, Kunming, China
- School of Life Science, Yunnan University, Kunming, China
| | - Yang Yu
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Yong Shen
- College of Agriculture and Biotechnology, Yunnan Agricultural University, Kunming, China
| | - Qin Liu
- School of Landscape and Horticulture, Yunnan Vocational and Technical College of Agriculture, Kunming, China
| | - Zhiwei Zhao
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, The University of Texas Health Science Center at San Antonio (UT Health), San Antonio, TX, United States
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, The University of Texas Health Science Center at San Antonio (UT Health), San Antonio, TX, United States
| |
Collapse
|
340
|
Nishimon S, Nishimon M, Nishino S. Tasimelteon for treating non-24-h sleep-wake rhythm disorder. Expert Opin Pharmacother 2019; 20:1065-1073. [DOI: 10.1080/14656566.2019.1603293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Shohei Nishimon
- Sleep and Circadian Neurobiology Laboratory, Psychiatry and Behavior Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mari Nishimon
- Sleep and Circadian Neurobiology Laboratory, Psychiatry and Behavior Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Seiji Nishino
- Sleep and Circadian Neurobiology Laboratory, Psychiatry and Behavior Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
341
|
Tang H, Ma M, Wu Y, Deng M, Hu F, Almansoub H, Huang H, Wang D, Zhou L, Wei N, Man H, Lu Y, Liu D, Zhu L. Activation of MT2 receptor ameliorates dendritic abnormalities in Alzheimer's disease via C/EBPα/miR-125b pathway. Aging Cell 2019; 18:e12902. [PMID: 30706990 PMCID: PMC6413662 DOI: 10.1111/acel.12902] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/26/2018] [Accepted: 12/16/2018] [Indexed: 01/24/2023] Open
Abstract
Impairments of dendritic trees and spines have been found in many neurodegenerative diseases, including Alzheimer's disease (AD), in which the deficits of melatonin signal pathway were reported. Melatonin receptor 2 (MT2) is widely expressed in the hippocampus and mediates the biological functions of melatonin. It is known that melatonin application is protective to dendritic abnormalities in AD. However, whether MT2 is involved in the neuroprotection and the underlying mechanisms are not clear. Here, we first found that MT2 is dramatically reduced in the dendritic compartment upon the insult of oligomer Aβ. MT2 activation prevented the Aβ-induced disruption of dendritic complexity and spine. Importantly, activation of MT2 decreased cAMP, which in turn inactivated transcriptional factor CCAAT/enhancer-binding protein α(C/EBPα) to suppress miR-125b expression and elevate the expression of its target, GluN2A. In addition, miR-125b mimics fully blocked the protective effects of MT2 activation on dendritic trees and spines. Finally, injection of a lentivirus containing a miR-125b sponge into the hippocampus of APP/PS1 mice effectively rescued the dendritic abnormalities and learning/memory impairments. Our data demonstrated that the cAMP-C/EBPα/miR-125b/GluN2A signaling pathway is important to the neuroprotective effects of MT2 activation in Aβ-induced dendritic injuries and learning/memory disorders, providing a novel therapeutic target for the treatment of AD synaptopathy.
Collapse
Affiliation(s)
- Hui Tang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Mei Ma
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Ying Wu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Man‐Fei Deng
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Fan Hu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Hasan.a.m.m. Almansoub
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - He‐Zhou Huang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Ding‐Qi Wang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Lan‐Ting Zhou
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Na Wei
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Department of Pathology, School of Basic MedicineZhengzhou UniversityZhengzhouChina
| | - Hengye Man
- Department of BiologyBoston UniversityBostonMassachusetts
| | - Youming Lu
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| | - Dan Liu
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
- Department of Genetics, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ling‐Qiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- The Institute of Brain Research, Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
342
|
Huang CC, Chiou CH, Liu SC, Hu SL, Su CM, Tsai CH, Tang CH. Melatonin attenuates TNF-α and IL-1β expression in synovial fibroblasts and diminishes cartilage degradation: Implications for the treatment of rheumatoid arthritis. J Pineal Res 2019; 66:e12560. [PMID: 30648758 DOI: 10.1111/jpi.12560] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/12/2018] [Accepted: 12/23/2018] [Indexed: 12/19/2022]
Abstract
The hormone melatonin has many properties, including antioxidant, anti-inflammatory, and immunomodulatory effects. Melatonin has been demonstrated to be beneficial in several inflammatory autoimmune diseases, but its effects in rheumatoid arthritis (RA) remain controversial. We sought to determine how melatonin regulates inflammation in RA. We found that melatonin dose-dependently inhibits tumor necrosis factor-α (TNF-α) and interleukin (IL)-1β expression through the PI3K/AKT, ERK, and NF-κB signaling pathways. We also identified that melatonin inhibits TNF-α and IL-1β production by upregulating miR-3150a-3p expression. Synovial tissue specimens from RA patients and culture of human rheumatoid fibroblast-like synoviocytes confirmed that the MT1 receptor is needed for the anti-inflammatory activities of melatonin. Importantly, melatonin also significantly reduced paw swelling, cartilage degradation, and bone erosion in the collagen-induced arthritis mouse model. Our results indicate that melatonin ameliorates RA by inhibiting TNF-α and IL-1β production through downregulation of the PI3K/AKT, ERK, NF-κB signaling pathways, as well as miR-3150a-3p overexpression. The role of melatonin as an adjuvant treatment in patients with RA deserves further clinical studies.
Collapse
Affiliation(s)
- Chien-Chung Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chen-Hsiang Chiou
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shan-Chi Liu
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Sung-Lin Hu
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Chun-Hao Tsai
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
343
|
Geng CA, Yang TH, Huang XY, Ma YB, Zhang XM, Chen JJ. Antidepressant potential of Uncaria rhynchophylla and its active flavanol, catechin, targeting melatonin receptors. JOURNAL OF ETHNOPHARMACOLOGY 2019; 232:39-46. [PMID: 30543912 DOI: 10.1016/j.jep.2018.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/02/2018] [Accepted: 12/08/2018] [Indexed: 05/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicines (TCMs) are fascinating sources for natural drug candidates. Uncaria rhynchophylla (Gouteng) is a famous TCM used for alleviating central nervous system (CNS) disorders, while its antidepressant constituents are still disputed. AIM OF THE STUDY The present study was designed to assess the antidepressant property of U. rhynchophylla and characterize the active constituents targeting melatonin receptors which are closely related to CNS diseases. MATERIALS AND METHODS The total extract and each fraction of U. rhynchophylla were extensively assessed for their agonistic activity on melatonin receptors in vitro. The following bioassay-guided fractionation yielded the active constituents, whose activity was confirmed by dose-dependent bioassay and antagonistic experiment on HEK293 cells. Their antidepressant effects were evaluated on forced swimming test (FST), tail suspension test (TST) and open-field test (OFT) mice models in vivo. Their metabolic profiles in mice plasma were analyzed by LCMS-IT-TOF. RESULTS The stems and hooks of U. rhynchophylla were revealed with agonistic activity on melatonin receptors (MT1 and MT2). Under the guidance of bioassay, two flavanols, catechin and epicatechin were obtained and showed obviously activity agitating MT1 (EC50 = 25.8 and 156.1 μM) and MT2 (EC50 = 47.3 and 208.8 μM) receptors. The agonistic activity of catechin on melatonin receptors can be antagonized by luzindole at the concentrations of 1.57-100 μM. Catechin could significantly reduce the immobility time in both FST and TST mice models at doses of 80 and 40 mg/kg, without obvious effect on locomotor activity in OFT mice model. Five phase II (M1-M5) and one phase I (M6) metabolites of catechin were detected in mice plasma after intragastric (i.g.) administration. CONCLUSION Catechin is a potent antidepressant candidate from U. rhynchophylla by targeting melatonin receptors. The main metabolic pathways of catechin in mice plasma are glucuronidation (M3) and methylated glucuronidation (M4 and M5). This study provides valuable information for understanding the antidepressant potency of Gouteng and its active constituents.
Collapse
Affiliation(s)
- Chang-An Geng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132# Lanhei Road, Kunming 650201, Yunnan, PR China; Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, PR China
| | - Tong-Hua Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132# Lanhei Road, Kunming 650201, Yunnan, PR China; Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, PR China
| | - Xiao-Yan Huang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132# Lanhei Road, Kunming 650201, Yunnan, PR China; Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, PR China
| | - Yun-Bao Ma
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132# Lanhei Road, Kunming 650201, Yunnan, PR China; Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, PR China
| | - Xue-Mei Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132# Lanhei Road, Kunming 650201, Yunnan, PR China; Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, PR China
| | - Ji-Jun Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132# Lanhei Road, Kunming 650201, Yunnan, PR China; Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
344
|
Chao CC, Chen PC, Chiou PC, Hsu CJ, Liu PI, Yang YC, Reiter R, Yang SF, Tang CH. Melatonin suppresses lung cancer metastasis by inhibition of epithelial–mesenchymal transition through targeting to Twist. Clin Sci (Lond) 2019; 133:709-722. [DOI: 10.1042/cs20180945] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
The epithelial–mesenchymal transition (EMT) phenotype, whereby mature epithelial cells undergo phenotype transition and differentiate into motile, invasive cells, has been indicated in tumor metastasis. The melatonin hormone secreted by the pineal gland has an antioxidant effect and protects cells against carcinogenic substances that reduce tumor progression. However, the effects of melatonin in EMT and lung cancer metastasis are largely unknown. We found that melatonin down-regulated EMT by inhibiting Twist/Twist1 (twist family bHLH transcription factor 1) expression. This effect was mediated by MT1 receptor, PLC, p38/ERK and β-catenin signaling cascades. Twist expression was positively correlated with tumor stage and negatively correlated with MT1 expression in lung cancer specimens. Furthermore, melatonin inhibited EMT marker expression and lung cancer metastasis to liver in vivo. Finally, melatonin shows promise in the treatment of lung cancer metastasis and deserves further study.
Collapse
Affiliation(s)
- Chia-Chia Chao
- Department of Respiratory Therapy, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Po-Chun Chen
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Pei-Chen Chiou
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chin-Jung Hsu
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Po-I Liu
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
- Department of Thoracic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Chen Yang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
345
|
Gao Y, Wu X, Zhao S, Zhang Y, Ma H, Yang Z, Yang W, Zhao C, Wang L, Zhang Q. Melatonin receptor depletion suppressed hCG-induced testosterone expression in mouse Leydig cells. Cell Mol Biol Lett 2019; 24:21. [PMID: 30915128 PMCID: PMC6416941 DOI: 10.1186/s11658-019-0147-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Melatonin receptors MT1 and MT2 (genes officially named MTNR1A and MTNR1B, respectively) play crucial roles in melatonin-mediated regulation of circadian rhythms, the immune system, and control of reproduction in seasonally breeding animals. In this study, immunolocalization assay showed that MT1 and MT2 are highly expressed in Leydig cell membrane. To understand the biological function of melatonin receptors in hCG-induced testosterone synthesis, we generated melatonin receptor knockdown cells using specific siRNA and performed testosterone detection after hCG treatment. We found that knockdown of melatonin receptors, especially MTNR1A, led to an obvious decrease (> 60%) of testosterone level. Our further study revealed that knockdown of melatonin receptors repressed expression, at both the mRNA level and the protein level, of key steroidogenic genes, such as p450scc, p450c17 and StAR, which are essential for testosterone synthesis. hCG triggered endoplasmic reticulum (ER) stress to regulate steroidogenic genes' expression and apoptosis. To further investigate the potential roles of melatonin receptors in hCG-induced regulation of ER stress and apoptosis, we examined expression of some crucial ER stress markers, including Grp78, Chop, ATF4, Xbp1, and IRE1. We found that inhibition of melatonin receptors increased hCG-induced expression of Grp78, Chop and ATF4, but not Xbp1 and IRE1, suggesting that hCG may modulate IRE1 signaling pathways in a melatonin receptor-dependent manner. In addition, our further data showed that knockdown of MTNR1A and MTNR1B promoted hCG-induced expression of apoptosis markers, including p53, caspase-3 and Bcl-2. These results suggested that the melatonin receptors MTNR1A and MTNR1B are essential to repress hCG-induced ER stress and cell apoptosis. Our studies demonstrated that the mammalian melatonin receptors MT1 and MT2 are involved in testosterone synthesis via mediating multiple cell pathways.
Collapse
Affiliation(s)
- Yuan Gao
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Xiaochun Wu
- 2College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu China
| | - Shuqin Zhao
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Yujun Zhang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Hailong Ma
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Zhen Yang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Wanghao Yang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Chen Zhao
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Li Wang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China
| | - Quanwei Zhang
- 1College of Life Science and Technology, Gansu Agricultural University, Lanzhou, 730070 Gansu China.,2College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu China
| |
Collapse
|
346
|
Grassi L, Nanni MG, Rodin G, Li M, Caruso R. The use of antidepressants in oncology: a review and practical tips for oncologists. Ann Oncol 2019; 29:101-111. [PMID: 29272358 DOI: 10.1093/annonc/mdx526] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The use of psychotropic drugs, namely those with an antidepressant profile (ADs), is a mandatory part of an integrated treatment of psychiatric disorders among cancer patients. We aimed to synthetize the most relevant data emerging from published studies to provide tips about the use of ADs in oncology. Design A search was made of the major databases over the last 30 years (Embase/Medline, PsycLIT, PsycINFO, the Cochrane Library), including narrative reviews, systematic reviews and meta-analyses summarizing the results from observational studies and randomized clinical trials assessing effectiveness, safety profile, interactions, contraindications and use of ADs in oncology with regard to both psychiatric (depressive spectrum, stress-related, anxiety disorders) and cancer-related symptoms (e.g. pain, hot flashes and fatigue). Results The weight of evidence supports the efficacy of ADs for more severe major depression in individuals with cancer and as an adjuvant treatment in cancer-related symptoms, although the methodological limitations of reported randomized controlled trials do not permit definite conclusions. Data also indicate that there should be caution in the use of ADs in cancer patients in terms of their safety profile and potential clinically significant interactions with other prescribed medications. Practical recommendations that have been made for the use of ADs in cancer patients, in the context of a multimodal approach to depression treatment, have been summarized here. Conclusions ADs are a relatively safe and effective treatment for more severe major depression in cancer patients. However, more research is urgently needed regarding the efficacy of ADs in different cancer types and cancer settings, their interactions with anticancer agents and their additive benefit when integrated with psychosocial interventions.
Collapse
Affiliation(s)
- L Grassi
- Department of Biomedical and Specialty Surgical Sciences, Institute of Psychiatry, University of Ferrara, Ferrara, Italy.,University Hospital Psychiatry Unit, Integrated Department of Mental Health and Addictive Behavior, S. Anna University Hospital and Health Authorities, Ferrara, Italy
| | - M G Nanni
- Department of Biomedical and Specialty Surgical Sciences, Institute of Psychiatry, University of Ferrara, Ferrara, Italy.,University Hospital Psychiatry Unit, Integrated Department of Mental Health and Addictive Behavior, S. Anna University Hospital and Health Authorities, Ferrara, Italy
| | - G Rodin
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Supportive Care, University Health Network, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - M Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Supportive Care, University Health Network, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - R Caruso
- Department of Biomedical and Specialty Surgical Sciences, Institute of Psychiatry, University of Ferrara, Ferrara, Italy.,University Hospital Psychiatry Unit, Integrated Department of Mental Health and Addictive Behavior, S. Anna University Hospital and Health Authorities, Ferrara, Italy
| |
Collapse
|
347
|
Xia Y, Chen S, Zeng S, Zhao Y, Zhu C, Deng B, Zhu G, Yin Y, Wang W, Hardeland R, Ren W. Melatonin in macrophage biology: Current understanding and future perspectives. J Pineal Res 2019; 66:e12547. [PMID: 30597604 DOI: 10.1111/jpi.12547] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/26/2018] [Accepted: 12/26/2018] [Indexed: 12/13/2022]
Abstract
Melatonin is a ubiquitous hormone found in various organisms and highly affects the function of immune cells. In this review, we summarize the current understanding of the significance of melatonin in macrophage biology and the beneficial effects of melatonin in macrophage-associated diseases. Enzymes associated with synthesis of melatonin, as well as membrane receptors for melatonin, are found in macrophages. Indeed, melatonin influences the phenotype polarization of macrophages. Mechanistically, the roles of melatonin in macrophages are related to several cellular signaling pathways, such as NF-κB, STATs, and NLRP3/caspase-1. Notably, miRNAs (eg, miR-155/-34a/-23a), cellular metabolic pathways (eg, α-KG, HIF-1α, and ROS), and mitochondrial dynamics and mitophagy are also involved. Thus, melatonin modulates the development and progression of various macrophage-associated diseases, such as cancer and rheumatoid arthritis. This review provides a better understanding about the importance of melatonin in macrophage biology and macrophage-associated diseases.
Collapse
Affiliation(s)
- Yaoyao Xia
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Siyuan Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Sijing Zeng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yuanyuan Zhao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Congrui Zhu
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoo Noses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Baichuan Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Guoqiang Zhu
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoo Noses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Laboratory of Animal Nutrition and Health and Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Wence Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| | - Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoo Noses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
348
|
Wang J, Jiang C, Zhang K, Lan X, Chen X, Zang W, Wang Z, Guan F, Zhu C, Yang X, Lu H, Wang J. Melatonin receptor activation provides cerebral protection after traumatic brain injury by mitigating oxidative stress and inflammation via the Nrf2 signaling pathway. Free Radic Biol Med 2019; 131:345-355. [PMID: 30553970 DOI: 10.1016/j.freeradbiomed.2018.12.014] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/09/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023]
Abstract
Traumatic brain injury (TBI) is a principal cause of death and disability worldwide. Melatonin, a hormone made by the pineal gland, is known to have anti-inflammatory and antioxidant properties. In this study, using a weight-drop model of TBI, we investigated the protective effects of ramelteon, a melatonin MT1/MT2 receptor agonist, and its underlying mechanisms of action. Administration of ramelteon (10 mg/kg) daily at 10:00 a.m. alleviated TBI-induced early brain damage on day 3 and long-term neurobehavioral deficits on day 28 in C57BL/6 mice. Ramelteon also increased the protein levels of interleukin (IL)-10, IL-4, superoxide dismutase (SOD), glutathione, and glutathione peroxidase and reduced the protein levels of IL-1β, tumor necrosis factor, and malondialdehyde in brain tissue and serum on days 1, 3, and 7 post-TBI. Similarly, ramelteon attenuated microglial and astrocyte activation in the perilesional cortex on day 3. Furthermore, ramelteon decreased Keap 1 expression, promoted nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear accumulation, and increased levels of downstream proteins, including SOD-1, heme oxygenase-1, and NQO1 on day 3 post-TBI. However, in Nrf2 knockout mice with TBI, ramelteon did not decrease the lesion volume, neuronal degeneration, or myelin loss on day 3; nor did it mitigate depression-like behavior or most motor behavior deficits on day 28. Thus, timed ramelteon treatment appears to prevent inflammation and oxidative stress via the Nrf2-antioxidant response element pathway and might represent a potential chronotherapeutic strategy for treating TBI.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Antioxidants/pharmacology
- Astrocytes/drug effects
- Astrocytes/metabolism
- Astrocytes/pathology
- Brain Edema/drug therapy
- Brain Edema/genetics
- Brain Edema/metabolism
- Brain Edema/pathology
- Brain Injuries, Traumatic/drug therapy
- Brain Injuries, Traumatic/genetics
- Brain Injuries, Traumatic/metabolism
- Brain Injuries, Traumatic/pathology
- Cerebral Cortex/drug effects
- Cerebral Cortex/metabolism
- Cerebral Cortex/pathology
- Disease Models, Animal
- Gene Expression Regulation
- Glutathione Peroxidase/genetics
- Glutathione Peroxidase/metabolism
- Indenes/pharmacology
- Inflammation
- Interleukin-10/genetics
- Interleukin-10/metabolism
- Interleukin-1beta/genetics
- Interleukin-1beta/metabolism
- Interleukin-4/genetics
- Interleukin-4/metabolism
- Kelch-Like ECH-Associated Protein 1/genetics
- Kelch-Like ECH-Associated Protein 1/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/drug effects
- Microglia/metabolism
- Microglia/pathology
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/metabolism
- Oxidative Stress/drug effects
- Receptor, Melatonin, MT1/agonists
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT1/metabolism
- Receptor, Melatonin, MT2/agonists
- Receptor, Melatonin, MT2/genetics
- Receptor, Melatonin, MT2/metabolism
- Signal Transduction
- Superoxide Dismutase/genetics
- Superoxide Dismutase/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Junmin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China; The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Weidong Zang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Zhongyu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Fangxia Guan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; School of Life Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Changlian Zhu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Göteborg 40530, Sweden
| | - Xiuli Yang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
349
|
|
350
|
Abstract
Despite considerable advances in the past few years, obesity and type 2 diabetes mellitus (T2DM) remain two major challenges for public health systems globally. In the past 9 years, genome-wide association studies (GWAS) have established a major role for genetic variation within the MTNR1B locus in regulating fasting plasma levels of glucose and in affecting the risk of T2DM. This discovery generated a major interest in the melatonergic system, in particular the melatonin MT2 receptor (which is encoded by MTNR1B). In this Review, we discuss the effect of melatonin and its receptors on glucose homeostasis, obesity and T2DM. Preclinical and clinical post-GWAS evidence of frequent and rare variants of the MTNR1B locus confirmed its importance in regulating glucose homeostasis and T2DM risk with minor effects on obesity. However, these studies did not solve the question of whether melatonin is beneficial or detrimental, an issue that will be discussed in the context of the peculiarities of the melatonergic system. Melatonin receptors might have therapeutic potential as they belong to the highly druggable G protein-coupled receptor superfamily. Clarifying the precise role of melatonin and its receptors on glucose homeostasis is urgent, as melatonin is widely used for other indications, either as a prescribed medication or as a supplement without medical prescription, in many countries in Europe and in the USA.
Collapse
Affiliation(s)
- Angeliki Karamitri
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
- Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | - Ralf Jockers
- Inserm, U1016, Institut Cochin, Paris, France.
- CNRS UMR 8104, Paris, France.
- Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France.
| |
Collapse
|