101
|
Endothelial dysfunction in individuals born after fetal growth restriction: cardiovascular and renal consequences and preventive approaches. J Dev Orig Health Dis 2017; 8:448-464. [PMID: 28460648 DOI: 10.1017/s2040174417000265] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Individuals born after intrauterine growth restriction (IUGR) have an increased risk of perinatal morbidity/mortality, and those who survive face long-term consequences such as cardiovascular-related diseases, including systemic hypertension, atherosclerosis, coronary heart disease and chronic kidney disease. In addition to the demonstrated long-term effects of decreased nephron endowment and hyperactivity of the hypothalamic-pituitary-adrenal axis, individuals born after IUGR also exhibit early alterations in vascular structure and function, which have been identified as key factors of the development of cardiovascular-related diseases. The endothelium plays a major role in maintaining vascular function and homeostasis. Therefore, it is not surprising that impaired endothelial function can lead to the long-term development of vascular-related diseases. Endothelial dysfunction, particularly impaired endothelium-dependent vasodilation and vascular remodeling, involves decreased nitric oxide (NO) bioavailability, impaired endothelial NO synthase functionality, increased oxidative stress, endothelial progenitor cells dysfunction and accelerated vascular senescence. Preventive approaches such as breastfeeding, supplementation with folate, vitamins, antioxidants, L-citrulline, L-arginine and treatment with NO modulators represent promising strategies for improving endothelial function, mitigating long-term outcomes and possibly preventing IUGR of vascular origin. Moreover, the identification of early biomarkers of endothelial dysfunction, especially epigenetic biomarkers, could allow early screening and follow-up of individuals at risk of developing cardiovascular and renal diseases, thus contributing to the development of preventive and therapeutic strategies to avert the long-term effects of endothelial dysfunction in infants born after IUGR.
Collapse
|
102
|
Ireland KE, Maloyan A, Myatt L. Melatonin Improves Mitochondrial Respiration in Syncytiotrophoblasts From Placentas of Obese Women. Reprod Sci 2017; 25:120-130. [PMID: 28443479 DOI: 10.1177/1933719117704908] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Maternal obesity is associated with increased oxidative stress but decreased placental mitochondrial respiration and expression of mitochondrial electron transport chain (ETC) complexes I to V. Melatonin acts as an antioxidant and prevents oxidative stress-induced changes in cytotrophoblasts. Placentas were collected at term by cesarean delivery from obese (first trimester body mass index [BMI] ≥30, n = 10) or lean (BMI < 25, n = 6) women. Cytotrophoblasts were isolated and allowed to syncytialize for 72 hours with or without melatonin (0.1-100 µM) for the last 24 hours. Mitochondrial respiratory parameters were measured in a Seahorse XF24. Expression of ETC complexes I to V and antioxidant enzymes was measured by Western blot. Maternal clinical characteristics of patients were similar except for BMI. No significant improvement in mitochondrial respiration occurred with addition of melatonin to trophoblasts of lean women. However, in trophoblasts from obese women, melatonin (10 and 100 µmol/L) significantly increased maximal respiration ( P = .01 and P = .009, respectively) and spare capacity ( P = .02 and P = .003, respectively) compared to the untreated control. No differences were detected in the expression of ETC complexes and superoxide dismutase 1 or 2 in trophoblasts treated with melatonin. The expression of glutathione peroxidase, which was significantly greater in trophoblast of obese compared to lean women ( P < .05), was decreased back to the level seen in trophoblast of lean women with addition of melatonin ( P = .02). Improved spare respiratory capacity, the cellular reserve, could impart a protective effect to the placenta and fetus in an adverse intrauterine environment or in response to additional stressors.
Collapse
Affiliation(s)
- Kayla E Ireland
- 1 Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Alina Maloyan
- 2 Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,3 Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Leslie Myatt
- 2 Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,4 Department of Obstetrics and Gynecology, Bob and Charlee Moore Institute for Nutrition and Wellness, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
103
|
Zheng X, Zhou J, Tan DX, Wang N, Wang L, Shan D, Kong J. Melatonin Improves Waterlogging Tolerance of Malus baccata (Linn.) Borkh. Seedlings by Maintaining Aerobic Respiration, Photosynthesis and ROS Migration. FRONTIERS IN PLANT SCIENCE 2017; 8:483. [PMID: 28424730 PMCID: PMC5380759 DOI: 10.3389/fpls.2017.00483] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 03/20/2017] [Indexed: 05/18/2023]
Abstract
Waterlogging, one of the notorious abiotic stressors, retards the growth of apple plants and reduces their production. Thus, it is an urgent agenda for scientists to identify the suitable remedies for this problem. In the current study, we found that melatonin significantly improved the tolerance of apple seedlings against waterlogging stress. This was indicated by the reduced chlorosis and wilting of the seedlings after melatonin applications either by leaf spray or root irrigation. The mechanisms involve in that melatonin functions to maintain aerobic respiration, preserves photosynthesis and reduces oxidative damage of the plants which are under waterlogging stress. Melatonin application also enhances the gene expression of its synthetic enzymes (MbT5H1, MbAANAT3, MbASMT9) and increases melatonin production. This is the first report of a positive feedback that exogenous melatonin application promotes the melatonin synthesis in plants. A post-transcriptional regulation apparently participated in this regulation. When exogenous melatonin meets the requirement of the plants it is found that the protein synthesis of MbASMT9 was suppressed. Taken together, the results showed that melatonin was an effective molecule to protect plant, particularly apple plant, against waterlogging stress.
Collapse
Affiliation(s)
- Xiaodong Zheng
- College of Horticulture, China Agricultural UniversityBeijing, China
| | - Jingzhe Zhou
- Beijing Soil and Fertilizer Work StationBeijing, China
| | - Dun-Xian Tan
- Department of Cellular and Structural Biology, UT Health Science Center San Antonio, San AntonioTX, USA
| | - Na Wang
- College of Horticulture, China Agricultural UniversityBeijing, China
| | - Lin Wang
- College of Horticulture, China Agricultural UniversityBeijing, China
| | - Dongqian Shan
- College of Horticulture, China Agricultural UniversityBeijing, China
| | - Jin Kong
- College of Horticulture, China Agricultural UniversityBeijing, China
| |
Collapse
|
104
|
Contribution of inducible and neuronal nitric oxide synthases to mitochondrial damage and melatonin rescue in LPS-treated mice. J Physiol Biochem 2017; 73:235-244. [PMID: 28110436 DOI: 10.1007/s13105-017-0548-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 01/16/2017] [Indexed: 12/14/2022]
Abstract
NOS isoform activation is related to liver failure during sepsis, but the mechanisms driving mitochondrial impairment remain unclear. We induced sepsis by LPS administration to inducible nitric oxide synthase (iNOS-/-) and neuronal nitric oxide synthase (nNOS-/-) mice and their respective wild-type controls to examine the contribution of iNOS to mitochondrial failure in the absence of nNOS. To achieve this goal, the determination of messenger RNA (mRNA) expression and protein content of iNOS in cytosol and mitochondria, the mitochondrial respiratory complex content, and the levels of nitrosative and oxidative stress (by measuring 3-nitrotyrosine residues and carbonyl groups, respectively) were examined in the liver of control and septic mice. We detected strongly elevated iNOS mRNA expression and protein levels in liver cytosol and mitochondria of septic mice, which were related to enhanced oxidative and nitrosative stress, and with fewer changes in respiratory complexes. The absence of the iNOS, but not nNOS, gene absolutely prevented mitochondrial impairment during sepsis. Moreover, the nNOS gene did not modify the expression and the effects of iNOS here shown. Melatonin administration counteracted iNOS activation and mitochondrial damage and enhanced the expression of the respiratory complexes above the control values. These effects were unrelated to the presence or absence of nNOS. iNOS is a main target to prevent liver mitochondrial impairment during sepsis, and melatonin represents an efficient antagonist of these iNOS-dependent effects whereas it may boost mitochondrial respiration to enhance liver survival.
Collapse
|
105
|
A novel combination treatment to stimulate bone healing and regeneration under hypoxic conditions: photobiomodulation and melatonin. Lasers Med Sci 2017; 32:533-541. [PMID: 28091848 DOI: 10.1007/s10103-017-2145-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 01/05/2017] [Indexed: 12/16/2022]
Abstract
Melatonin has anabolic effects on the bone, even under hypoxia, and laser irradiation has been shown to improve osteoblastic differentiation. The aim of this study was to investigate whether laser irradiation and melatonin would have synergistic effects on osteoblastic differentiation and mineralization under hypoxic conditions. MC3T3-E1 cells were exposed to 1% oxygen tension for the hypoxia condition. The cells were divided into four groups: G1-osteoblast differentiation medium only (as the hypoxic condition), G2-treatment with 50 μM melatonin only, G3-laser irradiation (808 nm, 80 mW, GaAlAs diode) only, and G4-treatment with 50 μM melatonin and laser irradiation (808 nm, 80 mW, GaAlAs diode). Immunoblotting showed that osterix expression was markedly increased in the melatonin-treated and laser-irradiated cells at 48 and 72 h. In addition, alkaline phosphatase activity significantly increased and continued to rise throughout the experiment. Alizarin Red staining showed markedly increased mineralized nodules as compared with only melatonin-treated or laser-irradiated cells at day 7, which significantly increased by day 14. Moreover, when melatonin-treated cells were laser-irradiated, the differentiation and mineralization of cells were found to involve p38 MAPK and PRKD1 signaling mechanisms. However, the enhanced effects of laser irradiation with melatonin were markedly inhibited when the cells were treated with luzindole, a selective melatonin receptor antagonist. Therefore, we concluded that laser irradiation could promote the effect of melatonin on the differentiation and mineralization of MC3T3-E1 cells under hypoxic conditions, and that this process is mediated through melatonin 1/2 receptors and PKRD/p38 signaling pathways.
Collapse
|
106
|
Ghareghani M, Dokoohaki S, Ghanbari A, Farhadi N, Zibara K, Khodadoust S, Parishani M, Ghavamizadeh M, Sadeghi H. Melatonin exacerbates acute experimental autoimmune encephalomyelitis by enhancing the serum levels of lactate: A potential biomarker of multiple sclerosis progression. Clin Exp Pharmacol Physiol 2016; 44:52-61. [DOI: 10.1111/1440-1681.12678] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/25/2016] [Accepted: 09/27/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Majid Ghareghani
- Cellular and Molecular Research Center; Faculty of Medicine; Yasuj University of Medical Sciences; Yasuj Iran
| | - Shima Dokoohaki
- Cellular and Molecular Research Center; Faculty of Medicine; Yasuj University of Medical Sciences; Yasuj Iran
| | - Amir Ghanbari
- Cellular and Molecular Research Center; Faculty of Medicine; Yasuj University of Medical Sciences; Yasuj Iran
| | - Naser Farhadi
- Cellular and Molecular Research Center; Faculty of Medicine; Yasuj University of Medical Sciences; Yasuj Iran
| | - Kazem Zibara
- ER045; Laboratory of Stem Cells; EDST; Biology Department; Faculty of Sciences; Lebanese University; Beirut Lebanon
| | - Saeid Khodadoust
- Department of Chemistry; Behbahan Khatam Alanbia University of Technology; Behbahan Iran
| | - Mohammad Parishani
- Cellular and Molecular Research Center; Faculty of Medicine; Yasuj University of Medical Sciences; Yasuj Iran
| | - Mehdi Ghavamizadeh
- Cellular and Molecular Gerash Research center; Gerash University of Medical Science; Gerash Iran
| | - Heibatollah Sadeghi
- Cellular and Molecular Research Center; Faculty of Medicine; Yasuj University of Medical Sciences; Yasuj Iran
| |
Collapse
|
107
|
Tan DX, Manchester LC, Qin L, Reiter RJ. Melatonin: A Mitochondrial Targeting Molecule Involving Mitochondrial Protection and Dynamics. Int J Mol Sci 2016; 17:ijms17122124. [PMID: 27999288 PMCID: PMC5187924 DOI: 10.3390/ijms17122124] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/28/2016] [Accepted: 12/07/2016] [Indexed: 01/19/2023] Open
Abstract
Melatonin has been speculated to be mainly synthesized by mitochondria. This speculation is supported by the recent discovery that aralkylamine N-acetyltransferase/serotonin N-acetyltransferase (AANAT/SNAT) is localized in mitochondria of oocytes and the isolated mitochondria generate melatonin. We have also speculated that melatonin is a mitochondria-targeted antioxidant. It accumulates in mitochondria with high concentration against a concentration gradient. This is probably achieved by an active transportation via mitochondrial melatonin transporter(s). Melatonin protects mitochondria by scavenging reactive oxygen species (ROS), inhibiting the mitochondrial permeability transition pore (MPTP), and activating uncoupling proteins (UCPs). Thus, melatonin maintains the optimal mitochondrial membrane potential and preserves mitochondrial functions. In addition, mitochondrial biogenesis and dynamics is also regulated by melatonin. In most cases, melatonin reduces mitochondrial fission and elevates their fusion. Mitochondrial dynamics exhibit an oscillatory pattern which matches the melatonin circadian secretory rhythm in pinealeocytes and probably in other cells. Recently, melatonin has been found to promote mitophagy and improve homeostasis of mitochondria.
Collapse
Affiliation(s)
- Dun-Xian Tan
- Department of Cell System and Anatomy, The University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | - Lucien C Manchester
- Department of Cell System and Anatomy, The University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | - Lilan Qin
- Department of Cell System and Anatomy, The University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | - Russel J Reiter
- Department of Cell System and Anatomy, The University of Texas Health Science Center, San Antonio, TX 78229, USA.
| |
Collapse
|
108
|
Phytanic Acid-Induced Neurotoxicological Manifestations and Apoptosis Ameliorated by Mitochondria-Mediated Actions of Melatonin. Mol Neurobiol 2016; 54:6960-6969. [PMID: 27785753 DOI: 10.1007/s12035-016-0209-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/11/2016] [Indexed: 12/27/2022]
Abstract
Phytanic acid, a saturated branched chain fatty acid and a major constituent of human diet, is predominantly found in dairy products, meat, and fish. It is a degradation product from the phytol side chain of chlorophyll. Degradation of PA is known to occur mainly in peroxisomes via α-oxidation and in mitochondria via β-oxidation. Due to its β-methyl group present at the 3-position of the carbon atoms, PA cannot be β-oxidized. Although alteration in the metabolism of PA may play an important role in neurodegeneration, the exact mechanism behind it remains to be evaluated. In this study, we have described the potential of PA to induce neurotoxicity as an in vitro model (neuronal cell line, SH-SY5Y cells). Cells were pretreated with melatonin (10 μM) for 1 h followed by with and without PA (100 μM) for 24 h. In the present study, our data has confirmed that PA markedly increased both intracellular reactive oxygen species and reactive nitrogen species levels. Our results have shown that PA treatment did not induce cell death by cleavage of caspase-3/PARP-1 mediated by mitochondria through intrinsic pathways; however, PA induced nitric oxide-dependent apoptosis in SH-SY5Y cells. Additionally, melatonin pretreatment reduced the cell death in SH-SY5Y cells. Melatonin also effectively exerted an antiapoptotic and anti-inflammatory action by regulating Bax, Bcl-2, p-NFκB, and iNOS expressions in SH-SY5Y cells. These results suggested that melatonin acted as an antioxidative and antiapoptotic agent by modulating ROS, apoptotic proteins, and inflammatory responses under BCFA-induced neurotoxic conditions. The protective effects of melatonin depend on direct scavenging activity of free radicals and indirect antioxidant effects. Further deciphering of the cellular and molecular mechanism associated with neuroprotection by melatonin is warranted in BCFA-induced neurotoxicity.
Collapse
|
109
|
Krityakiarana W, Sompup K, Jongkamonwiwat N, Mukda S, Pinilla FG, Govitrapong P, Phansuwan-Pujito P. Effects of melatonin on severe crush spinal cord injury-induced reactive astrocyte and scar formation. J Neurosci Res 2016; 94:1451-1459. [PMID: 27717042 DOI: 10.1002/jnr.23930] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023]
Abstract
The present work aimed at analyzing the effects of melatonin on scar formation after spinal cord injury (SCI). Upregulation of reactive astrocyte under SCI pathological conditions has been presented in several studies. It has been proved that the crucial factor in triggering this upregulation is proinflammatory cytokines. Moreover, scar formation is an important barrier to axonal regeneration through the lesion area. Melatonin plays an important role in reducing inflammation, but its effects on scar formation in the injured spinal cord remain unknown. Hence, we used the model of severe crush injury in mice to investigate the effects of melatonin on scar formation. Mice were randomly separated into four groups; SCI, SCI+Melatonin 1 (single dose), SCI+Melatonin 14 (14 daily doses), and control. Melatonin was administered by intraperitoneal injection (10 mg/kg) after injury. Immunohistochemical analysis, Western blot, and behavioral evaluation were used to explore the effects of melatonin after SCI for 14 days. The melatonin-treated mice presented higher expression of neuronal markers (P < 0.001). Remarkably, the inflammatory response appeared to be greatly reduced in the SCI+Melatonin 14 group (P < 0.001), which also displayed less scar formation (P < 0.05). These findings suggest that melatonin inhibits scar formation by acting on inflammatory cytokines after SCI. Overall, our results suggest that melatonin is a promising treatment strategy after SCI that deserves further investigation. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Warin Krityakiarana
- Division of Physical Therapy, Faculty of Health Science, Srinakharinwirot University, Nakhon-Nayok, Thailand. .,Department of Rehabilitation for Persons with Disabilities, Ratchasuda College, Mahidol University, Nakhonpathom, Thailand.
| | - Kamonrapat Sompup
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, Thailand
| | - Nopporn Jongkamonwiwat
- Division of Physical Therapy, Faculty of Health Science, Srinakharinwirot University, Nakhon-Nayok, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, Thailand
| | - Fernando Gomez Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, Thailand.,Center for Neuroscience and Department for Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
110
|
Díaz-Casado ME, Lima E, García JA, Doerrier C, Aranda P, Sayed RK, Guerra-Librero A, Escames G, López LC, Acuña-Castroviejo D. Melatonin rescues zebrafish embryos from the parkinsonian phenotype restoring the parkin/PINK1/DJ-1/MUL1 network. J Pineal Res 2016; 61:96-107. [PMID: 27064726 DOI: 10.1111/jpi.12332] [Citation(s) in RCA: 63] [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: 03/03/2016] [Accepted: 04/08/2016] [Indexed: 02/06/2023]
Abstract
Multiple studies reporting mitochondrial impairment in Parkinson's disease (PD) involve knockout or knockdown models to inhibit the expression of mitochondrial-related genes, including parkin, PINK1, and DJ-1 ones. Melatonin has significant neuroprotective properties, which have been related to its ability to boost mitochondrial bioenergetics. The meaning and molecular targets of melatonin in PD are yet unclear. Zebrafish are an outstanding model of PD because they are vertebrates, their dopaminergic system is comparable to the nigrostriatal system of humans, and their brains express the same genes as mammals. The exposure of 24 hpf zebrafish embryos to MPTP leads to a significant inhibition of the mitochondrial complex I and the induction of sncga gene, responsible for enhancing γ-synuclein accumulation, which is related to mitochondrial dysfunction. Moreover, MPTP inhibited the parkin/PINK1/DJ-1 expression, impeding the normal function of the parkin/PINK1/DJ-1/MUL1 network to remove the damaged mitochondria. This situation remains over time, and removing MPTP from the treatment did not stop the neurodegenerative process. On the contrary, mitochondria become worse during the next 2 days without MPTP, and the embryos developed a severe motor impairment that cannot be rescued because the mitochondrial-related gene expression remained inhibited. Melatonin, added together with MPTP or added once MPTP was removed, prevented and recovered, respectively, the parkinsonian phenotype once it was established, restoring gene expression and normal function of the parkin/PINK1/DJ-1/MUL1 loop and also the normal motor activity of the embryos. The results show, for the first time, that melatonin restores brain function in zebrafish suffering with Parkinson-like disease.
Collapse
Affiliation(s)
- María E Díaz-Casado
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Elena Lima
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - José A García
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Carolina Doerrier
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Paula Aranda
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Ramy Ka Sayed
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Ana Guerra-Librero
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Germaine Escames
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Luis C López
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Darío Acuña-Castroviejo
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Unidad de Gestión Clínica de Laboratorios Clínicos, Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
111
|
Souza WL, Moraes EA, Costa JM, Sousa PH, Lopes Junior ES, Oliveira RP, Toniolli R. Efeito de diferentes concentrações de melatonina em espermatozoides de carneiros sobre estresse oxidativo após criopreservação. PESQUISA VETERINARIA BRASILEIRA 2016. [DOI: 10.1590/s0100-736x2016000700017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Resumo: Objetivou-se avaliar o efeito da adição de diferentes concentrações de melatonina no sêmen diluído de carneiros após criopreservação. Foram coletados 10 ejaculados de três carneiros adultos (n=30), por meio de vagina artificial para ovinos. Os ejaculados coletados foram diluídos em Tris-Gema de ovo, para a concentração final de 200x106 sptz/mL, mantidos em banho maria a 32°C, e a melatonina adicionada conforme os tratamentos: Controle; 100pM; 100nM; 100μM e 1mM de melatonina. Então, as amostras foram resfriadas em câmara fria a 5°C por duas horas, envasadas em palhetas de 0,5 mL e lacradas. Logo após, foram acondicionadas sob vapores do nitrogênio liquido, por 15 minutos, a 8cm da lâmina líquida e congeladas com nitrogênio líquido. As amostras foram analisadas quanto à motilidade espermática, integridade da membrana plasmática, membrana acrossomal, atividade mitocondrial, quantificação do estresse oxidativo e a capacidade de ligação. As variáveis foram submetidas à análise de variância e as médias foram comparadas pelo teste de Tukey a 5% de probabilidade. A motilidade total e progressiva dos espermatozoides descongelados foi maior nas amostras tratadas com 100pM de melatonina (62,99 e 45,07% respectivamente; P<0,05) quando comparado aos demais tratamentos. A adição das diferentes concentrações de melatonina no sêmen diluído, com exceção da concentração de 1 mM, apresentou maior percentual de células com membrana plasmática íntegra, quando comparadas com o controle (P<0,05). O percentual de espermatozoides com integridade da membrana do acrossoma foi maior no sêmen tratado com 100 pM de melatonina (P<0,05) do que nos demais tratamentos. A alta atividade mitocondrial foi maior nos espermatozoides tratados com 100 pM de melatonina (69,30%; P<0,05). A adição de 100 nM de melatonina reduziu a quantidade de TBARS após a criopreservação (2,84; P<0,05) quando comparado aos demais tratamentos. Após o descongelamento, o número de espermatozoides que se ligaram à membrana perivitelina foi maior nos tratados com 100 pM de melatonina (155,73; P<0,05). Portanto, a adição de melatonina no sêmen diluído pode ser útil para aperfeiçoar a criopreservação do sêmen de ovinos, melhorando as taxas de fertilização por meio da inseminação artificial.
Collapse
|
112
|
Loureiro R, Magalhães-Novais S, Mesquita KA, Baldeiras I, Sousa IS, Tavares LC, Barbosa IA, Oliveira PJ, Vega-Naredo I. Melatonin antiproliferative effects require active mitochondrial function in embryonal carcinoma cells. Oncotarget 2016; 6:17081-96. [PMID: 26025920 PMCID: PMC4627293 DOI: 10.18632/oncotarget.4012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/08/2015] [Indexed: 12/28/2022] Open
Abstract
Although melatonin oncostatic and cytotoxic effects have been described in different types of cancer cells, the specific mechanisms leading to its antitumoral effects and their metabolic context specificity are still not completely understood. Here, we evaluated the effects of melatonin in P19 embryonal carcinoma stem cells (CSCs) and in their differentiated counterparts, cultured in either high glucose medium or in a galactose (glucose-free) medium which leads to glycolytic suppression and increased mitochondrial metabolism. We found that highly glycolytic P19 CSCs were less susceptible to melatonin antitumoral effects while cell populations relying on oxidative metabolism for ATP production were more affected. The observed antiproliferative action of melatonin was associated with an arrest at S-phase, decreased oxygen consumption, down-regulation of BCL-2 expression and an increase in oxidative stress culminating with caspase-3-independent cell death. Interestingly, the combined treatment of melatonin and dichloroacetate had a synergistic effect in cells grown in the galactose medium and resulted in an inhibitory effect in the highly resistant P19 CSCs. Melatonin appears to exert its antiproliferative activity in P19 carcinoma cells through a mitochondrially-mediated action which in turn allows the amplification of the effects of dichloroacetate, even in cells with a more glycolytic phenotype.
Collapse
Affiliation(s)
- Rute Loureiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Silvia Magalhães-Novais
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Katia A Mesquita
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ines Baldeiras
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,School of Medicine, University of Coimbra, Coimbra, Portugal
| | - Isabel S Sousa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ludgero C Tavares
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ines A Barbosa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ignacio Vega-Naredo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain
| |
Collapse
|
113
|
Pacini N, Borziani F. Oncostatic-Cytoprotective Effect of Melatonin and Other Bioactive Molecules: A Common Target in Mitochondrial Respiration. Int J Mol Sci 2016; 17:341. [PMID: 26959015 PMCID: PMC4813203 DOI: 10.3390/ijms17030341] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 11/27/2015] [Accepted: 12/03/2015] [Indexed: 12/27/2022] Open
Abstract
For several years, oncostatic and antiproliferative properties, as well as thoses of cell death induction through 5-methoxy-N-acetiltryptamine or melatonin treatment, have been known. Paradoxically, its remarkable scavenger, cytoprotective and anti-apoptotic characteristics in neurodegeneration models, such as Alzheimer’s disease and Parkinson’s disease are known too. Analogous results have been confirmed by a large literature to be associated to the use of many other bioactive molecules such as resveratrol, tocopherol derivatives or vitamin E and others. It is interesting to note that the two opposite situations, namely the neoplastic pathology and the neurodegeneration, are characterized by deep alterations of the metabolome, of mitochondrial function and of oxygen consumption, so that the oncostatic and cytoprotective action can find a potential rationalization because of the different metabolic and mitochondrial situations, and in the effect that these molecules exercise on the mitochondrial function. In this review we discuss historical and general aspects of melatonin, relations between cancers and the metabolome and between neurodegeneration and the metabolome, and the possible effects of melatonin and of other bioactive molecules on metabolic and mitochondrial dynamics. Finally, we suggest a common general mechanism as responsible for the oncostatic/cytoprotective effect of melatonin and of other molecules examined.
Collapse
Affiliation(s)
- Nicola Pacini
- Laboratorio Privato di Biochimica F. Pacini, via trabocchetto 10, 89126 Reggio Calabria, Italy.
| | - Fabio Borziani
- Laboratorio Privato di Biochimica F. Pacini, via trabocchetto 10, 89126 Reggio Calabria, Italy.
| |
Collapse
|
114
|
Pang YW, Sun YQ, Sun WJ, Du WH, Hao HS, Zhao SJ, Zhu HB. Melatonin inhibits paraquat-induced cell death in bovine preimplantation embryos. J Pineal Res 2016; 60:155-66. [PMID: 26607207 DOI: 10.1111/jpi.12297] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/19/2015] [Indexed: 12/20/2022]
Abstract
Preimplantation embryos are sensitive to oxidative stress-induced damage that can be caused by reactive oxygen species (ROS) originating from normal embryonic metabolism and/or the external surroundings. Paraquat (PQ), a commonly used pesticide and potent ROS generator, can induce embryotoxicity. The present study aimed to investigate the effects of melatonin on PQ-induced damage during embryonic development in bovine preimplantation embryos. PQ treatment significantly reduced the ability of bovine embryos to develop to the blastocyst stage, and the addition of melatonin markedly reversed the developmental failure caused by PQ (20.9% versus 14.3%). Apoptotic assay showed that melatonin pretreatment did not change the total cell number in blastocysts, but the incidence of apoptotic nuclei and the release of cytochrome c were significantly decreased. Using real-time quantitative polymerase chain reaction analysis, we found that melatonin pre-incubation significantly altered the expression levels of genes associated with redox signaling, particularly by attenuating the transcript level of Txnip and reinforcing the expression of Trx. Furthermore, melatonin pretreatment significantly reduced the expression of the pro-apoptotic caspase-3 and Bax, while the expression of the anti-apoptotic Bcl-2 and XIAP was unaffected. Western blot analysis showed that melatonin protected bovine embryos from PQ-induced damage in a p38-dependent manner, but extracellular signal-regulated kinase (ERK) and c-JUN N-terminal kinase (JNK) did not appear to be involved. Together, these results identify an underlying mechanism by which melatonin enhances the developmental potential of bovine preimplantation embryos under oxidative stress conditions.
Collapse
Affiliation(s)
- Yun-Wei Pang
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ye-Qing Sun
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wei-Jun Sun
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wei-Hua Du
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hai-Sheng Hao
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shan-Jiang Zhao
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hua-Bin Zhu
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
115
|
Volt H, García JA, Doerrier C, Díaz-Casado ME, Guerra-Librero A, López LC, Escames G, Tresguerres JA, Acuña-Castroviejo D. Same molecule but different expression: aging and sepsis trigger NLRP3 inflammasome activation, a target of melatonin. J Pineal Res 2016; 60:193-205. [PMID: 26681113 DOI: 10.1111/jpi.12303] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/11/2015] [Indexed: 12/13/2022]
Abstract
The connection between the innate immune system, clock genes, and mitochondrial bioenergetics was analyzed during aging and sepsis in mouse heart. Our results suggest that the sole NF-κB activation does not explain the inflammatory process underlying aging; the former also triggers the NLRP3 inflammasome that enhances caspase-1-dependent maturation of IL-1β. In this way, aged mice enter into a vicious cycle as IL-1β further activates the NF-κB/NLRP3 inflammasome link. The origin of NF-κB activation was related to the age-dependent Bmal1/Clock/RORα/Rev-Erbα loop disruption, which lowers NAD(+) levels, reducing the SIRT1 deacetylase ability to inactivate NF-κB. Consequently, NF-κB binding to DNA increases, raising the formation of proinflammatory mediators and inducing mitochondrial impairment. The cycle is then closed with the subsequent NLRP3 inflammasome activation. This paired contribution of the innate immune pathways serves as a catalyst to magnify the response to sepsis in aged compared with young mice. Melatonin administration blunted the septic response, reducing inflammation and oxidative stress, and enhancing mitochondrial function at the levels of nonseptic aged mice, but it did not counteract the age-related inflammation. Together, our results suggest that, although with different strengths, chronoinflammaging constitutes the biochemical substrate of aging and sepsis, and identifies the NLRP3 inflammasome as a new molecular target for melatonin, providing a rationale for its use in NLRP3-dependent diseases.
Collapse
Affiliation(s)
- Huayqui Volt
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - José A García
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Carolina Doerrier
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - María E Díaz-Casado
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Ana Guerra-Librero
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Luis C López
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Germaine Escames
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Jesús A Tresguerres
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Darío Acuña-Castroviejo
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
116
|
Adikwu E, Nelson B, Atuboyedia Obianime W. Beneficial effects of melatonin and alpha lipoic acid on lopinavir/ ritonavir-induced alterations in serum lipid and glucose levels of male albino rats. MAKEDONSKO FARMACEVTSKI BILTEN 2016. [DOI: 10.33320/maced.pharm.bull.2016.62.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The use of lopinavir/ritonavir (LPV/r) has been associated with alterations in serum lipid and glucose levels. This study was designed to investigate the effects of melatonin (MT) and alpha lipoic acid (ALA) on LPV/r-induced changes in serum levels of triglycerides (TG), total cholesterol (TC), low density lipoprotein cholesterol (LDL-C), very low density lipoprotein cholesterol (VLDL-C) and glucose (GL) levels in male albino rats. Rats in group A1 (placebo control) and group A2 (solvent control) were orally treated with normal saline and 1% ethanol respectively. Groups B-F contained 15 rats each which were divided into 3 groups of 5 rats each. Rats in group B were orally treated with MT (10 mg kg-1/day), ALA (10 mg kg-1/day) and a combination of MT and ALA, accordingly. Rats in group C were orally treated with 22.9/5.71, 45.6/11.4 and 91.4/22.9 mg kg-1/day of LPV/r, accordingly. Rats in groups D-F were orally treated with MT (10 mg kg-1/ day), ALA (10 mg kg-1/day) and combined doses of ALA and MT before oral treatment with 22.9/5.71, 45.6/11.4 and 91.4/22.9 mg kg-1/day of LPV/r, accordingly. All rats were treated for 30 days and at the end of the drug treatment, the serum levels of lipid fractions and glucose were evaluated. Treatment with MT and ALA significantly (p<0.05) decreased baseline serum levels of TG, TC, VLDL-C, LDL-C and GL, but these parameters were significantly (p<0.05) increased in a dose-dependent manner in LPV/r-treated rats when compared to placebo control. Administration of MT and ALA prior to treatment with LPV/r significantly (p<0.05) decreased serum levels of TG, TC, VLDL-C, LDL-C and GL when compared to LPV/r-treated rats. However, decreases obtained in rats pretreated with combined doses of MT and ALA were significantly (p<0.05) different when compared to their individual doses. This study showed that MT and ALA can serve as remedies for LPV/r-induced alterations in serum lipid and glucose levels.
Collapse
|
117
|
Berger HR, Morken TS, Vettukattil R, Brubakk AM, Sonnewald U, Widerøe M. No improvement of neuronal metabolism in the reperfusion phase with melatonin treatment after hypoxic-ischemic brain injury in the neonatal rat. J Neurochem 2015; 136:339-50. [PMID: 26526584 DOI: 10.1111/jnc.13420] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/22/2015] [Accepted: 10/23/2015] [Indexed: 01/13/2023]
Abstract
Mitochondrial impairment is a key feature underlying neonatal hypoxic-ischemic (HI) brain injury and melatonin is potentially neuroprotective through its effects on mitochondria. In this study, we have used (1) H and (13) C NMR spectroscopy after injection of [1-(13) C]glucose and [1,2-(13) C]acetate to examine neuronal and astrocytic metabolism in the early reperfusion phase after unilateral HI brain injury in 7-day-old rat pups, exploring the effects of HI on mitochondrial function and the potential protective effects of melatonin on brain metabolism. One hour after hypoxia-ischemia, astrocytic metabolism was recovered and glycolysis was normalized, whereas mitochondrial metabolism in neurons was clearly impaired. Pyruvate carboxylation was also lower in both hemispheres after HI. The transfer of glutamate from neurons to astrocytes was higher whereas the transfer of glutamine from astrocytes to neurons was lower 1 h after HI in the contralateral hemisphere. Neuronal metabolism was equally affected in pups treated with melatonin (10 mg/kg) immediately after HI as in vehicle treated pups indicating that the given dose of melatonin was not capable of protecting the neuronal mitochondria in this early phase after HI brain injury. However, any beneficial effects of melatonin might have been masked by modulatory effects of the solvent dimethyl sulfoxide on cerebral metabolism. Neuronal and astrocytic metabolism was examined by (13) C and (1) H NMR spectroscopy in the early reperfusion phase after unilateral hypoxic-ischemic brain injury and melatonin treatment in neonatal rats. One hour after hypoxia-ischemia astrocytic mitochondrial metabolism had recovered and glycolysis was normalized, whereas mitochondrial metabolism in neurons was impaired. Melatonin treatment did not show a protective effect on neuronal metabolism.
Collapse
Affiliation(s)
- Hester R Berger
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tora Sund Morken
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Ophthalmology, Trondheim University Hospital, Trondheim, Norway
| | - Riyas Vettukattil
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ann-Mari Brubakk
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ursula Sonnewald
- Department of Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marius Widerøe
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
118
|
Solanas E, Sostres C, Serrablo A, García-Gil A, García JJ, Aranguren FJ, Jiménez P, Hughes RD, Serrano MT. Effect of Dimethyl Sulfoxide and Melatonin on the Isolation of Human Primary Hepatocytes. Cells Tissues Organs 2015; 200:316-25. [DOI: 10.1159/000433521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2015] [Indexed: 11/19/2022] Open
|
119
|
Agil A, El-Hammadi M, Jiménez-Aranda A, Tassi M, Abdo W, Fernández-Vázquez G, Reiter RJ. Melatonin reduces hepatic mitochondrial dysfunction in diabetic obese rats. J Pineal Res 2015; 59:70-9. [PMID: 25904243 DOI: 10.1111/jpi.12241] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/20/2015] [Indexed: 12/20/2022]
Abstract
Hepatic mitochondrial dysfunction is thought to play a role in the development of liver steatosis and insulin resistance, which are both common characteristics of obesity and type 2 diabetes mellitus (T2DM). It was hypothesized that the antioxidant properties of melatonin could potentially improve the impaired functions of hepatic mitochondria in diabetic obese animals. Male Zucker diabetic fatty (ZDF) rats and lean littermates (ZL) were given either melatonin (10 mg/kg BW/day) orally for 6 wk (M-ZDF and M-ZL) or vehicle as control groups (C-ZDF and C-ZL). Hepatic function was evaluated by measurement of serum alanine transaminase and aspartate transaminase levels, liver histopathology and electron microscopy, and hepatic mitochondrial functions. Several impaired functions of hepatic mitochondria were observed in C-ZDF in comparison with C-ZL rats. Melatonin treatment to ZDF rats decreases serum levels of ALT (P < 0.001), alleviates liver steatosis and vacuolation, and also mitigates diabetic-induced mitochondrial abnormalities, glycogen, and lipid accumulation. Melatonin improves mitochondrial dysfunction in M-ZDF rats by increasing activities of mitochondrial citrate synthase (P < 0.001) and complex IV of electron transfer chain (P < 0.05) and enhances state 3 respiration (P < 0.001), respiratory control index (RCR) (P < 0.01), and phosphorylation coefficient (ADP/O ratio) (P < 0.05). Also melatonin augments ATP production (P < 0.05) and diminishes uncoupling protein 2 levels (P < 0.001). These results demonstrate that chronic oral melatonin reduces liver steatosis and mitochondria dysfunction in ZDF rats. Therefore, it may be beneficial in the treatment of diabesity.
Collapse
Affiliation(s)
- Ahmad Agil
- Department of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, Granada, Spain
| | - Mazen El-Hammadi
- Department of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, Granada, Spain
- Department of Pharmaceutics, Faculty of Pharmacy, Damascus University, Damascus, Syria
| | - Aroa Jiménez-Aranda
- Department of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, Granada, Spain
| | - Mohamed Tassi
- Service of Microscopy, CIBM, University of Granada, Granada, Spain
| | - Walied Abdo
- Department of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, Granada, Spain
- Department of pathology, Faculty of Veterinary medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | | | - Russel J Reiter
- Department of cellular and Structural Biology, University of Texas Health Science at San Antonio, San Antonio, TX, USA
| |
Collapse
|
120
|
Thakor AS, Allison BJ, Niu Y, Botting KJ, Serón-Ferré M, Herrera EA, Giussani DA. Melatonin modulates the fetal cardiovascular defense response to acute hypoxia. J Pineal Res 2015; 59:80-90. [PMID: 25908097 PMCID: PMC4528231 DOI: 10.1111/jpi.12242] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 04/20/2015] [Indexed: 12/12/2022]
Abstract
Experimental studies in animal models supporting protective effects on the fetus of melatonin in adverse pregnancy have prompted clinical trials in human pregnancy complicated by fetal growth restriction. However, the effects of melatonin on the fetal defense to acute hypoxia, such as that which may occur during labor, remain unknown. This translational study tested the hypothesis, in vivo, that melatonin modulates the fetal cardiometabolic defense responses to acute hypoxia in chronically instrumented late gestation fetal sheep via alterations in fetal nitric oxide (NO) bioavailability. Under anesthesia, 6 fetal sheep at 0.85 gestation were instrumented with vascular catheters and a Transonic flow probe around a femoral artery. Five days later, fetuses were exposed to acute hypoxia with or without melatonin treatment. Fetal blood was taken to determine blood gas and metabolic status and plasma catecholamine concentrations. Hypoxia during melatonin treatment was repeated during in vivo NO blockade with the NO clamp. This technique permits blockade of de novo synthesis of NO while compensating for the tonic production of the gas, thereby maintaining basal cardiovascular function. Melatonin suppressed the redistribution of blood flow away from peripheral circulations and the glycemic and plasma catecholamine responses to acute hypoxia. These are important components of the fetal brain sparing response to acute hypoxia. The effects of melatonin involved NO-dependent mechanisms as the responses were reverted by fetal treatment with the NO clamp. Melatonin modulates the in vivo fetal cardiometabolic responses to acute hypoxia by increasing NO bioavailability.
Collapse
Affiliation(s)
- Avnesh S Thakor
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Beth J Allison
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Kimberley J Botting
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - Maria Serón-Ferré
- Facultad de Medicina, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Emilio A Herrera
- Facultad de Medicina, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Dino A Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
121
|
Prophylactic melatonin significantly reduces Alzheimer's neuropathology and associated cognitive deficits independent of antioxidant pathways in AβPP(swe)/PS1 mice. Mol Neurodegener 2015; 10:27. [PMID: 26159703 PMCID: PMC4702331 DOI: 10.1186/s13024-015-0027-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/30/2015] [Indexed: 02/06/2023] Open
Abstract
Background Alzheimer’s disease (AD) underlies dementia for millions of people worldwide, and its occurrence is set to double in the next 20 years. Currently, approved drugs for treating AD only marginally ameliorate cognitive deficits, and provide limited symptomatic relief, while newer substances under therapeutic development are potentially years away from benefiting patients. Melatonin (MEL) for insomnia has been proven safe with >15 years of over-the-counter access in the US. MEL exerts multiple complementary mechanisms of action against AD in animal models; thus it may be an excellent disease-modifying therapeutic. While presumed to provide neuroprotection via activation of known G-protein-coupled melatonin receptors (MTNRs), some data indicate MEL acts intracellularly to protect mitochondria and neurons by scavenging reactive oxygen species and reducing free radical formation. We examined whether genetic deletion of MTNRs abolishes MEL’s neuroprotective actions in the AβPPswe/PSEN1dE9 mouse model of AD (2xAD). Beginning at 4 months of age, both AD and control mice either with or without both MTNRs were administered either MEL or vehicle in drinking water for 12 months. Results Behavioral and cognitive assessments of 15-month-old AD mice revealed receptor-dependent effects of MEL on spatial learning and memory (Barnes maze, Morris Water Maze), but receptor-independent neuroprotective actions of MEL on non-spatial cognitive performance (Novel Object Recognition Test). Similarly, amyloid plaque loads in hippocampus and frontal cortex, as well as plasma Aβ1–42 levels, were significantly reduced by MEL in a receptor-independent manner, in contrast to MEL’s efficacy in reducing cortical antioxidant gene expression (Catalase, SOD1, Glutathione Peroxidase-1, Nrf2) only when receptors were present. Increased cytochrome c oxidase activity was seen in 16mo AD mice as compared to non-AD control mice. This increase was completely prevented by MEL treatment of 2xAD/MTNR+ mice, but only partially prevented in 2xAD/MTNR- mice, consistent with mixed receptor-dependent and independent effects of MEL on this measure of mitochondrial function. Conclusions These findings demonstrate that prophylactic MEL significantly reduces AD neuropathology and associated cognitive deficits in a manner that is independent of antioxidant pathways. Future identification of direct molecular targets for MEL action in the brain should open new vistas for development of better AD therapeutics.
Collapse
|
122
|
|
123
|
Krestinina OV, Baburina YL, Azarashvili TS. Effect of melatonin on stress-induced opening of non-selective pore in mitochondria from brain of young and old rats. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2015. [DOI: 10.1134/s1990747814020032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
124
|
García JA, Volt H, Venegas C, Doerrier C, Escames G, López LC, Acuña-Castroviejo D. Disruption of the NF-κB/NLRP3 connection by melatonin requires retinoid-related orphan receptor-α and blocks the septic response in mice. FASEB J 2015; 29:3863-75. [PMID: 26045547 DOI: 10.1096/fj.15-273656] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/18/2015] [Indexed: 01/06/2023]
Abstract
We determined the NF-κB- and NOD-like receptor (NLR)P3-dependent molecular mechanisms involved in sepsis and evaluated the role of retinoid-related orphan receptor (ROR)-α in melatonin's anti-inflammatory actions. Western blot, RT-PCR, ELISA, and spectrophotometric analysis revealed that NF-κB and NLRP3 closely interact, leading to proinflammatory and pro-oxidant status in heart tissue of septic C57BL/6J mice. Moreover, mitochondrial oxygen consumption was reduced by 80% in septic mice. In vivo and in vitro analysis showed that melatonin administration blunts NF-κB transcriptional activity through a sirtuin1-dependent NF-κB deacetylation in septic mice. Melatonin also decreased NF-κB-dependent proinflammatory response and restored redox balance and mitochondrial homeostasis, thus inhibiting the NLRP3 inflammasome. In an important finding, the inhibition of NF-κB by melatonin, but not that of NLRP3, was blunted in RORα (sg/sg) mice, indicating that functional RORα transcription factor is necessary for the initiation of the innate immune response against inflammation. Our results are evidence of the NF-κB/NLRP3 connection during sepsis and identify NLRP3 as a novel molecular target for melatonin. The multiple molecular targets of melatonin in this study explain its potent anti-inflammatory efficacy against systemic innate immune activation and herald a promising therapeutic application for melatonin in the treatment of sepsis.
Collapse
Affiliation(s)
- José A García
- *Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, and Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain; and Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| | - Huayqui Volt
- *Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, and Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain; and Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| | - Carmen Venegas
- *Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, and Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain; and Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| | - Carolina Doerrier
- *Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, and Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain; and Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| | - Germaine Escames
- *Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, and Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain; and Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| | - Luis C López
- *Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, and Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain; and Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| | - Darío Acuña-Castroviejo
- *Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, and Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain; and Unidad de Gestión Clínica de Laboratorios, Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
125
|
Torres F, González-Candia A, Montt C, Ebensperger G, Chubretovic M, Serón-Ferré M, Reyes RV, Llanos AJ, Herrera EA. Melatonin reduces oxidative stress and improves vascular function in pulmonary hypertensive newborn sheep. J Pineal Res 2015; 58:362-73. [PMID: 25736256 DOI: 10.1111/jpi.12222] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 02/26/2015] [Indexed: 12/12/2022]
Abstract
Pulmonary hypertension of the newborn (PHN) constitutes a critical condition with severe cardiovascular and neurological consequences. One of its main causes is hypoxia during gestation, and thus, it is a public health concern in populations living above 2500 m. Although some mechanisms are recognized, the pathophysiological facts that lead to PHN are not fully understood, which explains the lack of an effective treatment. Oxidative stress is one of the proposed mechanisms inducing pulmonary vascular dysfunction and PHN. Therefore, we assessed whether melatonin, a potent antioxidant, improves pulmonary vascular function. Twelve newborn sheep were gestated, born, and raised at 3600 meters. At 3 days old, lambs were catheterized and daily cardiovascular measurements were recorded. Lambs were divided into two groups, one received daily vehicle as control and another received daily melatonin (1 mg/kg/d), for 8 days. At 11 days old, lung tissue and small pulmonary arteries (SPA) were collected. Melatonin decreased pulmonary pressure and resistance for the first 3 days of treatment. Further, melatonin significantly improved the vasodilator function of SPA, enhancing the endothelial- and muscular-dependent pathways. This was associated with an enhanced nitric oxide-dependent and nitric oxide independent vasodilator components and with increased nitric oxide bioavailability in lung tissue. Further, melatonin reduced the pulmonary oxidative stress markers and increased enzymatic and nonenzymatic antioxidant capacity. Finally, these effects were associated with an increase of lumen diameter and a mild decrease in the wall of the pulmonary arteries. These outcomes support the use of melatonin as an adjuvant in the treatment for PHN.
Collapse
Affiliation(s)
- Flavio Torres
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Providencia, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Hardeland R, Cardinali DP, Brown GM, Pandi-Perumal SR. Melatonin and brain inflammaging. Prog Neurobiol 2015; 127-128:46-63. [DOI: 10.1016/j.pneurobio.2015.02.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 10/27/2014] [Accepted: 02/05/2015] [Indexed: 02/07/2023]
|
127
|
Effect of seasons on semen production, effect of melatonin on the liquid storage (5 °C) with correlated study of birth rate in mithun (Bos frontalis). ASIAN PACIFIC JOURNAL OF REPRODUCTION 2015. [DOI: 10.1016/s2305-0500(14)60049-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
128
|
Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Protective role of melatonin in mitochondrial dysfunction and related disorders. Arch Toxicol 2015; 89:923-39. [DOI: 10.1007/s00204-015-1475-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
|
129
|
Solanas E, Sostres C, Serrablo A, García-Gil A, Aranguren F, Jimenez P, Serrano MT. Incubation with dimethyl sulfoxide prior to cryopreservation improves functionality of thawed human primary hepatocytes. Biopreserv Biobank 2015; 10:446-53. [PMID: 24845046 DOI: 10.1089/bio.2012.0015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Efficient cryopreservation of human hepatocytes is essential for their use in cell therapy. This study investigated the effects of adding melatonin and/or dimethyl sulfoxide (DMSO) to pre-incubation and/or freezing solutions on the viability and function of thawed human hepatocytes. METHODS Isolated human hepatocytes were pre-incubated for 90 min at 4°C in Williams' Medium E (WEM), WEM containing 5 mM melatonin dissolved in DMSO, or WEM containing the equivalent amount of DMSO (1%). The hepatocytes were frozen in University of Wisconsin solution (UW) and 10% DMSO, with or without 5 mM melatonin. After thawing, viability, plating efficiency, mitochondrial dehydrogenase activity (MTT), and albumin and urea production were analyzed. RESULTS Viability and plating efficiency were not affected by melatonin or DMSO in pre-incubation media. Unexpectedly, hepatocytes pre-incubated with DMSO had significantly higher MTT (29.7% vs. control, p<0.01), albumin (82.8% vs. control, p<0.05), and urea amounts (26.2% vs. control, p=0.06) than those incubated only with WEM. Hepatocytes pre-incubated in media containing melatonin had amounts between those of cells incubated with DMSO or only with WEM (p<0.05 for MTT and p>0.05 for albumin and urea values). Also, the addition of melatonin to the freezing media did not significantly improve any of the studied parameters (p>0.05). DISCUSSION Adding 1% DMSO to pre-incubation media prior to the cryopreservation of human hepatocytes preserves hepatocyte function after thawing. These findings could be considered in current hepatocyte cryopreservation protocols.
Collapse
Affiliation(s)
- Estela Solanas
- 1 IIS Aragón, CIBER Enfermedades Hepáticas y Digestivas (CIBERehd). Molecular Research Laboratory , Zaragoza, Spain
| | | | | | | | | | | | | |
Collapse
|
130
|
Doerrier C, García JA, Volt H, Díaz-Casado ME, Lima-Cabello E, Ortiz F, Luna-Sánchez M, Escames G, López LC, Acuña-Castroviejo D. Identification of mitochondrial deficits and melatonin targets in liver of septic mice by high-resolution respirometry. Life Sci 2015; 121:158-65. [DOI: 10.1016/j.lfs.2014.11.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/11/2014] [Accepted: 11/21/2014] [Indexed: 11/15/2022]
|
131
|
Tanabe M, Tamura H, Taketani T, Okada M, Lee L, Tamura I, Maekawa R, Asada H, Yamagata Y, Sugino N. Melatonin protects the integrity of granulosa cells by reducing oxidative stress in nuclei, mitochondria, and plasma membranes in mice. J Reprod Dev 2014; 61:35-41. [PMID: 25366368 PMCID: PMC4354229 DOI: 10.1262/jrd.2014-105] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Melatonin protects luteinized granulosa cells (GCs) from oxidative stress in the follicle during ovulation. However, it is unclear in which cellular components (e.g., nuclei, mitochondria, or plasma membranes) melatonin works as an antioxidant. GCs from immature (3 wks) ICR mice were incubated with hydrogen peroxide (H2O2; 0.01, 0.1, 1, 10 mM) in the presence or absence of melatonin (100 μg/ml) for 2 h. DNA damage was assessed by fluorescence-based immunocytochemistry using specific antibodies for 8-hydroxydeoxyguanosine (8-OHdG), an indicator of oxidative guanine base damage in DNA, and for histone H2AX phosphorylation (γH2AX), a marker of double-strand breaks of DNA. Mitochondrial function was assessed by the fluorescence intensity of MitoTracker Red probes, which diffuse across the membrane and accumulate in mitochondria with active membrane potentials. Lipid peroxidation of plasma membranes was analyzed by measuring hexanoyl-lysine (HEL), a oxidative stress marker
for lipid peroxidation. Apoptosis of GCs was assessed by nuclear fragmentation using DAPI staining, and apoptotic activities were evaluated by caspase-3/7 activities. H2O2 treatment significantly increased the fluorescence intensities of 8-OHdG and γH2AX, reduced the intensity of MitoTracker Red in the mitochondria, increased HEL concentrations in GCs, and enhanced the number of apoptotic cells and caspase-3/7 activities. All these changes were significantly decreased by melatonin treatment. Melatonin reduced oxidative stress-induced DNA damage, mitochondrial dysfunction, lipid peroxidation, and apoptosis in GCs, suggesting that melatonin protects GCs by reducing oxidative stress of cellular components including nuclei, mitochondria, and plasma membranes. Melatonin helps to maintain the integrity of GCs as an antioxidant in the preovulatory follicle.
Collapse
Affiliation(s)
- Manabu Tanabe
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Zhang HM, Zhang Y. Melatonin: a well-documented antioxidant with conditional pro-oxidant actions. J Pineal Res 2014; 57:131-46. [PMID: 25060102 DOI: 10.1111/jpi.12162] [Citation(s) in RCA: 606] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/18/2014] [Indexed: 12/19/2022]
Abstract
Melatonin (N-acetyl-5-methoxytryptamine), an indoleamine produced in many organs including the pineal gland, was initially characterized as a hormone primarily involved in circadian regulation of physiological and neuroendocrine function. Subsequent studies found that melatonin and its metabolic derivatives possess strong free radical scavenging properties. These metabolites are potent antioxidants against both ROS (reactive oxygen species) and RNS (reactive nitrogen species). The mechanisms by which melatonin and its metabolites protect against free radicals and oxidative stress include direct scavenging of radicals and radical products, induction of the expression of antioxidant enzymes, reduction of the activation of pro-oxidant enzymes, and maintenance of mitochondrial homeostasis. In both in vitro and in vivo studies, melatonin has been shown to reduce oxidative damage to lipids, proteins and DNA under a very wide set of conditions where toxic derivatives of oxygen are known to be produced. Although the vast majority of studies proved the antioxidant capacity of melatonin and its derivatives, a few studies using cultured cells found that melatonin promoted the generation of ROS at pharmacological concentrations (μm to mm range) in several tumor and nontumor cells; thus, melatonin functioned as a conditional pro-oxidant. Mechanistically, melatonin may stimulate ROS production through its interaction with calmodulin. Also, melatonin may interact with mitochondrial complex III or mitochondrial transition pore to promote ROS production. Whether melatonin functions as a pro-oxidant under in vivo conditions is not well documented; thus, whether the reported in vitro pro-oxidant actions come into play in live organisms remains to be established.
Collapse
Affiliation(s)
- Hong-Mei Zhang
- Department of Clinical Oncology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | | |
Collapse
|
133
|
Teodoro BG, Baraldi FG, Sampaio IH, Bomfim LHM, Queiroz AL, Passos MA, Carneiro EM, Alberici LC, Gomis R, Amaral FG, Cipolla-Neto J, Araújo MB, Lima T, Akira Uyemura S, Silveira LR, Vieira E. Melatonin prevents mitochondrial dysfunction and insulin resistance in rat skeletal muscle. J Pineal Res 2014; 57:155-67. [PMID: 24981026 DOI: 10.1111/jpi.12157] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/27/2014] [Indexed: 01/02/2023]
Abstract
Melatonin has a number of beneficial metabolic actions and reduced levels of melatonin may contribute to type 2 diabetes. The present study investigated the metabolic pathways involved in the effects of melatonin on mitochondrial function and insulin resistance in rat skeletal muscle. The effect of melatonin was tested both in vitro in isolated rats skeletal muscle cells and in vivo using pinealectomized rats (PNX). Insulin resistance was induced in vitro by treating primary rat skeletal muscle cells with palmitic acid for 24 hr. Insulin-stimulated glucose uptake was reduced by palmitic acid followed by decreased phosphorylation of AKT which was prevented my melatonin. Palmitic acid reduced mitochondrial respiration, genes involved in mitochondrial biogenesis and the levels of tricarboxylic acid cycle intermediates whereas melatonin counteracted all these parameters in insulin-resistant cells. Melatonin treatment increases CAMKII and p-CREB but had no effect on p-AMPK. Silencing of CREB protein by siRNA reduced mitochondrial respiration mimicking the effect of palmitic acid and prevented melatonin-induced increase in p-AKT in palmitic acid-treated cells. PNX rats exhibited mild glucose intolerance, decreased energy expenditure and decreased p-AKT, mitochondrial respiration, and p-CREB and PGC-1 alpha levels in skeletal muscle which were restored by melatonin treatment in PNX rats. In summary, we showed that melatonin could prevent mitochondrial dysfunction and insulin resistance via activation of CREB-PGC-1 alpha pathway. Thus, the present work shows that melatonin play an important role in skeletal muscle mitochondrial function which could explain some of the beneficial effects of melatonin in insulin resistance states.
Collapse
Affiliation(s)
- Bruno G Teodoro
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of Sao Paulo (USP), Ribeirão Preto, Brazil; Federal Institute of Science Education and Technology of São Paulo, Sao Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Navarro-Alarcón M, Ruiz-Ojeda FJ, Blanca-Herrera RM, A-Serrano MM, Acuña-Castroviejo D, Fernández-Vázquez G, Agil A. Melatonin and metabolic regulation: a review. Food Funct 2014; 5:2806-32. [DOI: 10.1039/c4fo00317a] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
135
|
Jimenéz-Aranda A, Fernández-Vázquez G, Mohammad A-Serrano M, Reiter RJ, Agil A. Melatonin improves mitochondrial function in inguinal white adipose tissue of Zücker diabetic fatty rats. J Pineal Res 2014; 57:103-9. [PMID: 24867433 DOI: 10.1111/jpi.12147] [Citation(s) in RCA: 52] [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: 04/08/2014] [Accepted: 05/23/2014] [Indexed: 12/19/2022]
Abstract
Mitochondrial dysfunction in adipose tissue may contribute to obesity-related metabolic derangements such as type 2 diabetes mellitus (T2DM). Because mitochondria are a target for melatonin action, the goal of this study was to investigate the effects of melatonin on mitochondrial function in white (WAT) and beige inguinal adipose tissue of Zücker diabetic fatty (ZDF) rats, a model of obesity-related T2DM. In this experimental model, melatonin reduces obesity and improves the metabolic profile. At 6 wk of age, ZDF rats and lean littermates (ZL) were subdivided into two groups, each composed of four rats: control (C-ZDF and C-ZL) and treated with oral melatonin in the drinking water (10 mg/kg/day) for 6 wk (M-ZDF and M-ZL). After the treatment period, animals were sacrificed, tissues dissected, and mitochondrial function assessed in isolated organelles. Melatonin increased the respiratory control ratio (RCR) in mitochondria from white fat of both lean (by 26.5%, P < 0.01) and obese (by 34.5%, P < 0.01) rats mainly through a reduction of proton leaking component of respiration (state 4) (28% decrease in ZL, P < 0.01 and 35% in ZDF, P < 0.01). However, melatonin treatment lowered the RCR in beige mitochondria of both lean (by 7%, P < 0.05) and obese (by 13%, P < 0.05) rats by maintaining high rates of uncoupled respiration. Melatonin also lowered mitochondrial oxidative status by reducing nitrite levels and by increasing superoxide dismutase activity. Moreover, melatonin treatment also caused a profound inhibition of Ca-induced opening of mPTP in isolated mitochondria from both types of fat, white and beige, in both lean and obese rats. These results demonstrate that chronic oral melatonin improves mitochondrial respiration and reduces the oxidative status and susceptibility to apoptosis in white and beige adipocytes. These melatonin effects help to prevent mitochondrial dysfunction and thereby to improve obesity-related metabolic disorders such as diabetes and dyslipidemia of ZDF rats.
Collapse
Affiliation(s)
- Aroa Jimenéz-Aranda
- Department of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, Granada, Spain
| | | | | | | | | |
Collapse
|
136
|
Elmahallawy EK, Jiménez-Aranda A, Martínez AS, Rodriguez-Granger J, Navarro-Alarcón M, Gutiérrez-Fernández J, Agil A. Activity of melatonin against Leishmania infantum promastigotes by mitochondrial dependent pathway. Chem Biol Interact 2014; 220:84-93. [PMID: 24973643 DOI: 10.1016/j.cbi.2014.06.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 06/06/2014] [Accepted: 06/16/2014] [Indexed: 10/25/2022]
Abstract
Visceral leishmaniasis, a potentially fatal disease, remains a major international health problem. Only a limited number of effective antileishmanial agents are available for chemotherapy, and many of them are expensive with severe side effects or have a markedly reduced effectiveness due to the development of drug resistance. Hence, there is a genuine need to develop a novel effective and less toxic antileishmanial drug. Melatonin, a neurohormone found in animals, plants, and microbes, can participate in various biological and physiological functions. Several in vitro or in vivo studies have reported the inhibitory effect of melatonin against many parasites via various mechanisms, including modulation of intracellular concentrations of calcium in the parasite and/or any other suggested mechanism. Importantly, many of available antileishmanial drugs have been reported to exert their effects by disrupting calcium homeostasis in the parasite. The objective of the present study was to test the efficacy of exogenous melatonin against Leishmania infantum promastigotes in vitro. Interestingly, melatonin not only demonstrated a significant antileishmanial activity of against promastigote viability in tested cultures but was also accompanied by an alteration of the calcium homeostasis of parasite mitochondrion, represented by earlier mitochondrial permeability transition pore opening, and by changes in some mitochondrial parameters are critical to parasite survival. These pioneering findings suggest that melatonin may be a candidate for the development of novel effective antileishmanial agents either alone or in associations with other drugs.
Collapse
Affiliation(s)
- Ehab Kotb Elmahallawy
- Department of Microbiology, Faculty of Medicine, University of Granada, Granada, Spain; Department of Zoonotic diseases, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Aroa Jiménez-Aranda
- Department of Pharmacology and Neurosciences Institute (CIBM), Faculty of Medicine, University of Granada, Granada, Spain
| | | | - Javier Rodriguez-Granger
- Service of Microbiology and Parasitology, University Hospital Virgen de las Nieves, Granada, Spain
| | - Miguel Navarro-Alarcón
- Department of Nutrition and Food Science, School of Pharmacy, University of Granada, Granada, Spain
| | | | - Ahmad Agil
- Department of Pharmacology and Neurosciences Institute (CIBM), Faculty of Medicine, University of Granada, Granada, Spain.
| |
Collapse
|
137
|
Bioactive compounds derived from the yeast metabolism of aromatic amino acids during alcoholic fermentation. BIOMED RESEARCH INTERNATIONAL 2014; 2014:898045. [PMID: 24895623 PMCID: PMC4026967 DOI: 10.1155/2014/898045] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/16/2014] [Indexed: 12/12/2022]
Abstract
Metabolites resulting from nitrogen metabolism in yeast are currently found in some fermented beverages such as wine and beer. Their study has recently attracted the attention of researchers. Some metabolites derived from aromatic amino acids are bioactive compounds that can behave as hormones or even mimic their role in humans and may also act as regulators in yeast. Although the metabolic pathways for their formation are well known, the physiological significance is still far from being understood. The understanding of this relevance will be a key element in managing the production of these compounds under controlled conditions, to offer fermented food with specific enrichment in these compounds or even to use the yeast as nutritional complements.
Collapse
|
138
|
Wong CS, Jow GM, Kaizaki A, Fan LW, Tien LT. Melatonin ameliorates brain injury induced by systemic lipopolysaccharide in neonatal rats. Neuroscience 2014; 267:147-56. [PMID: 24613717 DOI: 10.1016/j.neuroscience.2014.02.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/21/2014] [Accepted: 02/21/2014] [Indexed: 11/26/2022]
Abstract
Our previous study showed that lipopolysaccharide (LPS)-induced brain injury in the neonatal rat is associated with nitrosative and oxidative stress. The present study was conducted to examine whether melatonin, an endogenous molecule with antioxidant properties, reduces systemic LPS-induced nitrosative and oxidative damage in the neonatal rat brain. Intraperitoneal (i.p.) injection of LPS (2mg/kg) was administered to Sprague-Dawley rat pups on postnatal day 5 (P5), and i.p. administration of melatonin (20mg/kg) or vehicle was performed 5min after LPS injection. Sensorimotor behavioral tests were performed 24h after LPS exposure, and brain injury was examined after these tests. The results show that systemic LPS exposure resulted in impaired sensorimotor behavioral performance, and acute brain injury, as indicated by the loss of oligodendrocyte immunoreactivity and a decrease in mitochondrial activity in the neonatal rat brain. Melatonin treatment significantly reduced LPS-induced neurobehavioral disturbances and brain damage in neonatal rats. The neuroprotective effect of melatonin was associated with attenuation of LPS-induced nitrosative and oxidative stress, as indicated by the decreased nitrotyrosine- and 4-hydroxynonenal-positive staining in the brain following melatonin and LPS exposure in neonatal rats. Further, melatonin significantly attenuated LPS-induced increases in the number of activated microglia in the neonatal rat brain. The protection provided by melatonin was also associated with a reduced number of inducible nitric oxide synthase (iNOS)+ cells, which were double-labeled with ED1 (microglia). Our results show that melatonin prevents the brain injury and neurobehavioral disturbances induced by systemic LPS exposure in neonatal rats, and its neuroprotective effects are associated with its impact on nitrosative and oxidative stress.
Collapse
Affiliation(s)
- C-S Wong
- Department of Anesthesiology, Cathay General Hospital, Taipei City, Taiwan, ROC; School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan, ROC
| | - G-M Jow
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan, ROC
| | - A Kaizaki
- Department of Pharmacology, Toxicology and Therapeutics, Division of Toxicology, School of Pharmacy, Showa University, Shingawa-ku, Tokyo 142-8555, Japan
| | - L-W Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - L-T Tien
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan, ROC.
| |
Collapse
|
139
|
Martin-Cano FE, Camello-Almaraz C, Acuña-Castroviejo D, Pozo MJ, Camello PJ. Age-related changes in mitochondrial function of mouse colonic smooth muscle: beneficial effects of melatonin. J Pineal Res 2014; 56:163-74. [PMID: 24313280 DOI: 10.1111/jpi.12109] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 12/03/2013] [Indexed: 01/13/2023]
Abstract
Aging is a multifactorial process that involves biochemical, structural, and functional changes in mitochondria. The ability of melatonin to palliate the alterations induced by aging is based on its chronobiologic, antioxidant, and mitochondrial effects. There is little information about the effects of melatonin on the in situ mitochondrial network of aging cells and its physiological implications. We have studied the ability of melatonin to prevent the functional alterations of in situ mitochondria of smooth muscle cells and its impact on contractility. Mitochondrial membrane potential was recorded in isolated colonic smooth muscle cells from young mice (3 month old), aged mice (22-24-month old), and aged mice treated with melatonin (starting at 14-month age). Aging induced a partial mitochondrial depolarization in resting conditions and reduced the depolarizing response to cellular stimulation. Use of oligomycin indicated that aging enhanced the resting activity of the mitochondrial ATP synthase, whereas in young cells, the enzyme operated mainly in reverse mode. Melatonin treatment prevented all these changes. Aging reduced both spontaneous and stimulated contraction of colonic strips and shifted the metabolic dependence of contraction from mitochondria to glycolysis, as indicated the use of mitochondrial and glycolysis inhibitors. These functional alterations were also palliated by melatonin treatment. Aging effects were not related to a decrease in Ca2+ store mobilization, because this was enhanced in aged cells and restored by melatonin. In conclusion, melatonin prevents the age induced in situ mitochondrial potential alterations in smooth muscle cells and the associated changes in contractility and metabolism.
Collapse
Affiliation(s)
- Francisco E Martin-Cano
- Department of Physiology, Faculty of Nursing and Occupational Therapy, University of Extremadura, Cáceres, Spain
| | | | | | | | | |
Collapse
|
140
|
Park JH, Shim HM, Na AY, Bae KC, Bae JH, Im SS, Cho HC, Song DK. Melatonin prevents pancreatic β-cell loss due to glucotoxicity: the relationship between oxidative stress and endoplasmic reticulum stress. J Pineal Res 2014; 56:143-153. [PMID: 24168371 DOI: 10.1111/jpi.12106] [Citation(s) in RCA: 27] [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: 08/22/2013] [Accepted: 10/25/2013] [Indexed: 12/31/2022]
Abstract
Prolonged hyperglycemia results in pancreatic β-cell dysfunction and apoptosis, referred to as glucotoxicity. Although both oxidative and endoplasmic reticulum (ER) stresses have been implicated as major causative mechanisms of β-cell glucotoxicity, the reciprocal importance between the two remains to be elucidated. The aim of this study was to evaluate the differential effect of oxidative stress and ER stress on β-cell glucotoxicity, by employing melatonin which has free radical-scavenging and antioxidant properties. As expected, in β-cells exposed to prolonged high glucose levels, cell viability and glucose-stimulated insulin secretion (GSIS) were significantly impaired. Melatonin treatment markedly attenuated cellular apoptosis by scavenging reactive oxygen species via its plasmalemmal receptor-independent increase in antioxidant enzyme activity. However, treatments with antioxidants alone were insufficient to recover the impaired GSIS. Interestingly, 4-phenylbutyric acid (4-PBA), a chemical chaperone that attenuate ER stress by stabilizing protein structure, alleviated the impaired GSIS, but not apoptosis, suggesting that glucotoxicity induces oxidative and ER stress independently. We found that cotreatment of glucotoxic β-cells with melatonin and 4-PBA dramatically improved both their survival and insulin secretion. Taken together, these results suggest that ER stress may be the more critical mechanism for prolonged high-glucose-induced GSIS impairment, whereas oxidative stress appears to be more critical for the impaired β-cell viability. Therefore, combinatorial therapy of melatonin with an ER stress modifier may help recover pancreatic β-cells under glucotoxic conditions in type 2 diabetes.
Collapse
Affiliation(s)
- Jae-Hyung Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | | | | | | | | | | | | | | |
Collapse
|
141
|
Favero G, Rodella LF, Reiter RJ, Rezzani R. Melatonin and its atheroprotective effects: a review. Mol Cell Endocrinol 2014; 382:926-37. [PMID: 24291636 DOI: 10.1016/j.mce.2013.11.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/11/2013] [Accepted: 11/21/2013] [Indexed: 01/20/2023]
Abstract
Atherosclerosis is a chronic vascular disease in which oxidative stress and inflammation are commonly implicated as major causative factors. Identification of novel strategies that contribute to plaque stabilization or inhibition represents a continuing challenge for the medical community. The evidence from the last decade highlights that melatonin influences the cardiovascular system, but its mechanisms of action have not been definitively clarified. Melatonin has atheroprotective effects by acting on different pathogenic signaling processes; these result from its direct free radical scavenger activity, its indirect antioxidant properties and its anti-inflammatory actions. In this review, we summarize the many pieces of the puzzle which identified molecular targets for prevention and therapy against the atherosclerotic pathogenic processes and we evaluate the data documenting that melatonin treatment has important actions that protect against atherosclerosis and atherosclerosis-related cardiovascular diseases.
Collapse
Affiliation(s)
- Gaia Favero
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Luigi Fabrizio Rodella
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, USA
| | - Rita Rezzani
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| |
Collapse
|
142
|
Gao W, Lin M, Liang A, Zhang L, Chen C, Liang G, Xu C, Peng Y, Chen C, Huang D, Su P. Melatonin enhances chondrogenic differentiation of human mesenchymal stem cells. J Pineal Res 2014; 56:62-70. [PMID: 24117903 DOI: 10.1111/jpi.12098] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 09/20/2013] [Indexed: 01/30/2023]
Abstract
Intramembranous ossification and endochondral ossification are two ways through which bone formation and fracture healing occur. Accumulating amounts of evidence suggests that melatonin affects osteoblast differentiation, but little is known about the effects of melatonin on the process of chondrogenic differentiation. In this study, the effects of melatonin on human mesenchymal stem cells (MSCs) undergoing chondrogenic differentiation were investigated. Cells were induced along chondrogenic differentiation via high-density micromass culture in chondrogenic medium containing vehicle or 50 nm melatonin. Histological study and quantitative analysis of glycosaminoglycan (GAG) showed induced cartilage tissues to be larger and richer in GAG, collagen type II and collagen type X in the melatonin group than in the untreated controls. Real-time RT-PCR analysis demonstrated that melatonin treatment significantly up-regulated the expression of the genes involved in chondrogenic differentiation, including aggrecan (ACAN), collagen type II (COL2A1), collagen type X (COL10A1), SRY (sex-determining region Y)-box 9 (SOX9), runt-related transcription factor 2 (RUNX2) and the potent inducer of chondrogenic differentiation, bone morphogenetic protein 2 (BMP2). And the expression of melatonin membrane receptors (MT) MT1 and MT2 were detected in the chondrogenic-induced-MSCs by immunofluorescence staining. Luzindole, a melatonin receptor antagonist, was found to partially block the ability of melatonin to increase the size and GAG synthesis of the induced cartilage tissues, as well as to completely reverse the effect of melatonin on the gene expression of ACAN, COL2A1, COL10A1, SOX9 and BMP2 after 7 days of differentiation. These findings demonstrate that melatonin enhances chondrogenic differentiation of human MSCs at least partially through melatonin receptors.
Collapse
Affiliation(s)
- Wenjie Gao
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Ortiz F, García JA, Acuña-Castroviejo D, Doerrier C, López A, Venegas C, Volt H, Luna-Sánchez M, López LC, Escames G. The beneficial effects of melatonin against heart mitochondrial impairment during sepsis: inhibition of iNOS and preservation of nNOS. J Pineal Res 2014; 56:71-81. [PMID: 24117944 DOI: 10.1111/jpi.12099] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 09/20/2013] [Indexed: 01/17/2023]
Abstract
While it is accepted that the high production of nitric oxide (NO˙) by the inducible nitric oxide synthase (iNOS) impairs cardiac mitochondrial function during sepsis, the role of neuronal nitric oxide synthase (nNOS) may be protective. During sepsis, there is a significantly increase in the expression and activity of mitochondrial iNOS (i-mtNOS), which parallels the changes in cytosolic iNOS. The existence of a constitutive NOS form (c-mtNOS) in heart mitochondria has been also described, but its role in the heart failure during sepsis remains unclear. Herein, we analyzed the changes in mitochondrial oxidative stress and bioenergetics in wild-type and nNOS-deficient mice during sepsis, and the role of melatonin, a known antioxidant, in these changes. Sepsis was induced by cecal ligation and puncture, and heart mitochondria were analyzed for NOS expression and activity, nitrites, lipid peroxidation, glutathione and glutathione redox enzymes, oxidized proteins, and respiratory chain activity in vehicle- and melatonin-treated mice. Our data show that sepsis produced a similar induction of iNOS/i-mtNOS and comparable inhibition of the respiratory chain activity in wild-type and in nNOS-deficient mice. Sepsis also increased mitochondrial oxidative/nitrosative stress to a similar extent in both mice strains. Melatonin administration inhibited iNOS/i-mtNOS induction, restored mitochondrial homeostasis in septic mice, and preserved the activity of nNOS/c-mtNOS. The effects of melatonin were unrelated to the presence or the absence of nNOS. Our observations show a lack of effect of nNOS on heart bioenergetic impairment during sepsis and further support the beneficial actions of melatonin in sepsis.
Collapse
Affiliation(s)
- Francisco Ortiz
- Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Instituto de Biotecnología, Universidad de Granada, Granada, Spain; Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
SHE FEI, WANG WENBO, WANG YAN, TANG PEIFU, WEI JUNQIANG, CHEN HUA, ZHANG BOXUN. Melatonin protects MG63 osteoblast-like cells from hydrogen peroxide-induced cytotoxicity by maintaining mitochondrial function. Mol Med Rep 2013; 9:493-8. [DOI: 10.3892/mmr.2013.1832] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 11/18/2013] [Indexed: 11/06/2022] Open
|
145
|
Jiménez-Aranda A, Fernández-Vázquez G, Campos D, Tassi M, Velasco-Perez L, Tan DX, Reiter RJ, Agil A. Melatonin induces browning of inguinal white adipose tissue in Zucker diabetic fatty rats. J Pineal Res 2013; 55:416-23. [PMID: 24007241 DOI: 10.1111/jpi.12089] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 08/06/2013] [Indexed: 01/24/2023]
Abstract
Melatonin limits obesity in rodents without affecting food intake and activity, suggesting a thermogenic effect. Identification of brown fat (beige/brite) in white adipose tissue (WAT) prompted us to investigate whether melatonin is a brown-fat inducer. We used Zücker diabetic fatty (ZDF) rats, a model of obesity-related type 2 diabetes and a strain in which melatonin reduces obesity and improves their metabolic profiles. At 5 wk of age, ZDF rats and lean littermates (ZL) were subdivided into two groups, each composed of four rats: control and those treated with oral melatonin in the drinking water (10 mg/kg/day) for 6 wk. Melatonin induced browning of inguinal WAT in both ZDF and ZL rats. Hematoxylin-eosin staining showed patches of brown-like adipocytes in inguinal WAT in ZDF rats and also increased the amounts in ZL animals. Inguinal skin temperature was similar in untreated lean and obese rats. Melatonin increased inguinal temperature by 1.36 ± 0.02°C in ZL and by 0.55 ± 0.04°C in ZDF rats and sensitized the thermogenic effect of acute cold exposure in both groups. Melatonin increased the amounts of thermogenic proteins, uncoupling protein 1 (UCP1) (by ~2-fold, P < 0.01) and PGC-1α (by 25%, P < 0.05) in extracts from beige inguinal areas in ZL rats. Melatonin also induced measurable amounts of UCP1 and stimulated by ~2-fold the levels of PGC-1α in ZDF animals. Locomotor activity and circulating irisin levels were not affected by melatonin. These results demonstrate that chronic oral melatonin drives WAT into a brown-fat-like function in ZDF rats. This may contribute to melatonin's control of body weight and its metabolic benefits.
Collapse
Affiliation(s)
- Aroa Jiménez-Aranda
- Department of Pharmacology and Neurosciences Institute (CIBM), School of Medicine, University of Granada, Granada, Spain
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Hardeland R. Melatonin and the theories of aging: a critical appraisal of melatonin's role in antiaging mechanisms. J Pineal Res 2013; 55:325-56. [PMID: 24112071 DOI: 10.1111/jpi.12090] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 08/23/2013] [Indexed: 02/06/2023]
Abstract
The classic theories of aging such as the free radical theory, including its mitochondria-related versions, have largely focused on a few specific processes of senescence. Meanwhile, numerous interconnections have become apparent between age-dependent changes previously thought to proceed more or less independently. Increased damage by free radicals is not only linked to impairments of mitochondrial function, but also to inflammaging as it occurs during immune remodeling and by release of proinflammatory cytokines from mitotically arrested, DNA-damaged cells that exhibit the senescence-associated secretory phenotype (SASP). Among other effects, SASP can cause mutations in stem cells that reduce the capacity for tissue regeneration or, in worst case, lead to cancer stem cells. Oxidative stress has also been shown to promote telomere attrition. Moreover, damage by free radicals is connected to impaired circadian rhythmicity. Another nexus exists between cellular oscillators and metabolic sensing, in particular to the aging-suppressor SIRT1, which acts as an accessory clock protein. Melatonin, being a highly pleiotropic regulator molecule, interacts directly or indirectly with all the processes mentioned. These influences are critically reviewed, with emphasis on data from aged organisms and senescence-accelerated animals. The sometimes-controversial findings obtained either in a nongerontological context or in comparisons of tumor with nontumor cells are discussed in light of evidence obtained in senescent organisms. Although, in mammals, lifetime extension by melatonin has been rarely documented in a fully conclusive way, a support of healthy aging has been observed in rodents and is highly likely in humans.
Collapse
Affiliation(s)
- Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| |
Collapse
|
147
|
Phillipson OT. Management of the aging risk factor for Parkinson's disease. Neurobiol Aging 2013; 35:847-57. [PMID: 24246717 DOI: 10.1016/j.neurobiolaging.2013.10.073] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 01/12/2023]
Abstract
The aging risk factor for Parkinson's disease is described in terms of specific disease markers including mitochondrial and gene dysfunctions relevant to energy metabolism. This review details evidence for the ability of nutritional agents to manage these aging risk factors. The combination of alpha lipoic acid, acetyl-l-carnitine, coenzyme Q10, and melatonin supports energy metabolism via carbohydrate and fatty acid utilization, assists electron transport and adenosine triphosphate synthesis, counters oxidative and nitrosative stress, and raises defenses against protein misfolding, inflammatory stimuli, iron, and other endogenous or xenobiotic toxins. These effects are supported by gene expression via the antioxidant response element (ARE; Keap/Nrf2 pathway), and by peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1 alpha), a transcription coactivator, which regulates gene expression for energy metabolism and mitochondrial biogenesis, and maintains the structural integrity of mitochondria. The effectiveness and synergies of the combination against disease risks are discussed in relation to gene action, dopamine cell loss, and the accumulation and spread of pathology via misfolded alpha-synuclein. In addition there are potential synergies to support a neurorestorative role via glial derived neurotrophic factor expression.
Collapse
Affiliation(s)
- Oliver T Phillipson
- School of Medical Sciences, University of Bristol, University Walk, Bristol, UK.
| |
Collapse
|
148
|
Laothong U, Pinlaor P, Boonsiri P, Pairojkul C, Priprem A, Johns NP, Charoensuk L, Intuyod K, Pinlaor S. Melatonin inhibits cholangiocarcinoma and reduces liver injury in Opisthorchis viverrini-infected and N-nitrosodimethylamine-treated hamsters. J Pineal Res 2013; 55:257-66. [PMID: 23772655 DOI: 10.1111/jpi.12068] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/16/2013] [Indexed: 12/22/2022]
Abstract
The human liver fluke Opisthorchis viverrini infection and N-nitrosodimethylamine (NDMA) administration induce cholangiocarcinoma (CCA) and liver injury in hamsters. Melatonin protects against liver injury and reduces the alteration of mitochondrial structure, mitochondrial membrane potential, and mitochondrial pro- and anti-apoptotic pathways in various cancer types. To investigate the chemopreventive effect of melatonin on CCA genesis and liver injury, hamsters were treated with a combination of O. viverrini infection and NDMA concurrently administered with melatonin (10 mg/kg and 50 mg/kg) for 120 days. Melatonin treatment at 50 mg/kg caused a significant reduction in liver/body weight ratios and decreased tumor volumes leading to an increase in the survival of animals. In the tumorous tissues, the high-dose melatonin reduced DNA fragmentation and mitochondrial apoptosis by inducing anti-apoptotic protein (Bcl-2) in the mitochondrial fraction and down-regulating cytochrome c, pro-apoptotic protein (Bax), and caspase-3 in tumor cytosol. Moreover, a high-dose melatonin treatment significantly increased mitochondrial antioxidant enzymes and prevented mitochondrial ultrastructure changes in the tumor. Overall, melatonin has potent chemopreventive effects in inhibiting CCA genesis and also reduces liver injury in hamster CCA, which, in part, might involve in the suppression of CCA by reducing tumor mitochondria alteration.
Collapse
Affiliation(s)
- Umawadee Laothong
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | | | | | | | | | | | | | | |
Collapse
|
149
|
|
150
|
Feng YM, Jia YF, Su LY, Wang D, Lv L, Xu L, Yao YG. Decreased mitochondrial DNA copy number in the hippocampus and peripheral blood during opiate addiction is mediated by autophagy and can be salvaged by melatonin. Autophagy 2013; 9:1395-406. [PMID: 23800874 DOI: 10.4161/auto.25468] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Drug addiction is a chronic brain disease that is a serious social problem and causes enormous financial burden. Because mitochondrial abnormalities have been associated with opiate addiction, we examined the effect of morphine on mtDNA levels in rat and mouse models of addiction and in cultured cells. We found that mtDNA copy number was significantly reduced in the hippocampus and peripheral blood of morphine-addicted rats and mice compared with control animals. Concordantly, decreased mtDNA copy number and elevated mtDNA damage were observed in the peripheral blood from opiate-addicted patients, indicating detrimental effects of drug abuse and stress. In cultured rat pheochromocytoma (PC12) cells and mouse neurons, morphine treatment caused many mitochondrial defects, including a reduction in mtDNA copy number that was mediated by autophagy. Knockdown of the Atg7 gene was able to counteract the loss of mtDNA copy number induced by morphine. The mitochondria-targeted antioxidant melatonin restored mtDNA content and neuronal outgrowth and prevented the increase in autophagy upon morphine treatment. In mice, coadministration of melatonin with morphine ameliorated morphine-induced behavioral sensitization, analgesic tolerance and mtDNA content reduction. During drug withdrawal in opiate-addicted patients and improvement of protracted abstinence syndrome, we observed an increase of serum melatonin level. Taken together, our study indicates that opioid addiction is associated with mtDNA copy number reduction and neurostructural remodeling. These effects appear to be mediated by autophagy and can be salvaged by melatonin.
Collapse
Affiliation(s)
- Yue-Mei Feng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province; Kunming Institute of Zoology; Kunming, Yunnan China; University of Chinese Academy of Sciences; Beijing, China
| | | | | | | | | | | | | |
Collapse
|