1
|
Ji SM, Yoo H, Kim JI, Choi MJ, Cheon HG. Melatonin induces white-to-beige adipocyte transdifferentiation through melatonin receptor 1-mediated direct browning and indirect M2 polarization. Mol Cell Endocrinol 2025; 597:112439. [PMID: 39653309 DOI: 10.1016/j.mce.2024.112439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Previous studies have shown that melatonin induces adipocyte browning in vivo. However, the underlying mechanisms of melatonin action at the cellular level remain elusive. In this study, we investigated the mechanisms underlying melatonin-induced browning in 3T3-L1 adipocytes and RAW 264.7 macrophages. Melatonin caused the transdifferentiation of fully differentiated white adipocytes into beige adipocytes, which involves the activation of melatonin receptor 1, followed by increased phosphorylation of p38 MAPK and Akt. Both luzindole (LZ), a non-selective melatonin receptor antagonist, and selective melatonin receptor 1 knockdown attenuated the browning effects of melatonin. Melatonin also induced M2 polarization in RAW 264.7, involving the melatonin receptor 1-Src-STAT3/STAT6 phosphorylation signaling cascade. Melatonin-treated M2-conditioned medium (CM) contained increased levels of catecholamine (CA) and induced beige adipocytes when treated with differentiated 3T3-L1 white adipocytes. In vivo oral administration of melatonin to high-fat diet (HFD)-induced obese (DIO) mice reduced body weight, accompanied by increased expression of uncoupling protein-1 (UCP1) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in subcutaneous adipose tissues. Moreover, arginase-1 (Arg1) and mannose receptor C type-1 (MRC1) levels were markedly higher in the melatonin-treated groups, suggesting that melatonin induces adipose browning and M2 polarization in vivo. Collectively, melatonin-induced adipocyte browning appeared to be reflected by the sum of melatonin receptor 1-activated direct browning effects and indirect M2 polarization-mediated effects.
Collapse
MESH Headings
- Animals
- Melatonin/pharmacology
- Mice
- 3T3-L1 Cells
- Cell Transdifferentiation/drug effects
- Adipocytes, Beige/metabolism
- Adipocytes, Beige/drug effects
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- RAW 264.7 Cells
- Male
- Mice, Inbred C57BL
- Signal Transduction/drug effects
- Receptor, Melatonin, MT1/metabolism
- Receptor, Melatonin, MT1/genetics
- Cell Polarity/drug effects
- Macrophages/metabolism
- Macrophages/drug effects
- Diet, High-Fat
- Obesity/metabolism
- Obesity/pathology
- Uncoupling Protein 1/metabolism
- Uncoupling Protein 1/genetics
- Tryptamines
Collapse
Affiliation(s)
- Seong Mi Ji
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Hana Yoo
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Jea Il Kim
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, Republic of Korea
| | - Mi Jin Choi
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Hyae Gyeong Cheon
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, Republic of Korea.
| |
Collapse
|
2
|
Cano-Barquilla P, Jiménez-Ortega V, Fernández-Mateos P, Virto L, Maldonado Bautista E, Perez-Miguelsanz J, Esquifino AI. Daily Lipolysis Gene Expression in Male Rat Mesenteric Adipose Tissue: Obesity and Melatonin Effects. Int J Mol Sci 2025; 26:577. [PMID: 39859293 PMCID: PMC11765279 DOI: 10.3390/ijms26020577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Melatonin is involved in various functions such as the timing of circadian rhythms, energy metabolism, and body mass gain in experimental animals. However, its effects on adipose tissue lipid metabolism are still unclear. This study analyzes the effects of melatonin on the relative gene expression of lipolytic proteins in rat mesenteric adipose tissue and free fatty acid (FFA) and glycerol plasma levels of male Wistar rats fed a high-fat (HFD) or maintenance diet. Four experimental groups were established: control, obese, and control or obese plus 2.3 mg/kg/day of melatonin in tap water. After 11 weeks, animals were sacrificed at different times throughout a 24 h cycle, and mesenteric adipose tissue and plasma samples were collected and analyzed. Cgi58, Perilipin, and Dgat1 gene expression, as well as FFA and glycerol concentrations, showed rhythm patterns in the control group. HFD disrupted those rhythm patterns and increased FFA and glycerol concentrations during the dark photoperiod. In both melatonin-treated groups, almost all analyzed genes showed circadian patterns. Notably, melatonin significantly prevented the increase in FFA levels during the dark photoperiod in obese rats (obese group: ~1100 mM vs. obese + melatonin group: ~600 μM, similar to control levels). However, the rhythmic pattern observed in control animals was not sustained. According to our results, melatonin could regulate circadian gene transcription of mesenteric adipose tissue lipolysis proteins. The effect of melatonin on preventing elevated FFA plasma levels associated with high-fat diet intake warrants further investigation.
Collapse
Affiliation(s)
- Pilar Cano-Barquilla
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| | - Vanesa Jiménez-Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| | - Pilar Fernández-Mateos
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Leire Virto
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Anatomía y Embriología, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Estela Maldonado Bautista
- Departamento de Anatomía y Embriología, Faculta de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Juliana Perez-Miguelsanz
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
- Departamento de Anatomía y Embriología, Faculta de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Ana I. Esquifino
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28003 Madrid, Spain; (L.V.); (J.P.-M.)
| |
Collapse
|
3
|
Liu G, Su F, Zou X, Yang X, Tian L. Impact of an extended light regimen imposed during nursery period on the performance and lipid metabolism of weanling pigs. Anim Biosci 2025; 38:176-188. [PMID: 39483030 PMCID: PMC11725737 DOI: 10.5713/ab.24.0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/05/2024] [Accepted: 09/06/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE This study aimed to assess the impact of a prolonged photoperiod on the growth performance and lipid metabolism of weaned piglets. METHODS Twenty-four piglets weaned at 28 days of age were randomly dichotomized into two groups that were alternatively subjected to either long photoperiod (LP) group (16 L:8 D) or short photoperiod (SP) group (10 L:14 D) for 42days. Four replicates of three animals per replicates were used per experimental treatment. RESULTS Our results demonstrated that prolonged photoperiod increased piglet body weight, average daily weight gain (ADG), backfat thickness (BF), backfat index during the nursery period, and increased ADG, average daily feed intake (ADFI), and decreased the F/G of piglets during the experiment days 29 to 42. Meanwhile, we observed LP piglets' plasma melatonin, growth hormone and serotonin levels were decreased at 14 d and 42 d compared to SP piglets. Moreover, up-regulated mRNA or protein expression of PPARγ and CEBPα, and lower mRNA or protein expression of MTR1, ATGL, HSL, PPARα, and CPT1α, were observed in back subcutaneous fat of LP group compared with that of SP group. Significant increases were observed in the mRNA or protein contents of lipogenic genes, including C/EBPα, SREBP-1c, ACCα, and FAS, in the liver of LP piglets, whereas CPT1α and ACOX1 mRNA levels and PPARα and MTR1 protein expression were significantly downregulated in LP group compared to SP group. Extended photoperiod also increased lipid content in longissimus dorsi muscle that was associated with higher mRNA or protein levels of SREBP-1c, ACCα, FAS, Pref1, and LPL, decreased mRNA or protein contents of LeptinR, MTR1, HSL, and ACOX1. CONCLUSION Together, these findings suggest that there is an advantage, in terms of growth performance and fat deposition, in imposing a prolonged light program (16-h light/d) on nursery piglets to alleviate the negative aspects of weaning stress.
Collapse
Affiliation(s)
- Guangfan Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095,
China
| | - Fen Su
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095,
China
| | - Xingyue Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095,
China
| | - Xingming Yang
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095,
China
| | - Liang Tian
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095,
China
| |
Collapse
|
4
|
Osawa S, Kato H, Kemmoku D, Yamaguchi S, Jiang L, Tsuchiya Y, Takakura H, Izawa T. Exercise training-driven exosomal miRNA-323-5p activity suppresses adipogenic conversion of 3T3-L1 cells via the DUSP3/ERK pathway. Biochem Biophys Res Commun 2024; 734:150447. [PMID: 39083976 DOI: 10.1016/j.bbrc.2024.150447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Adipose-derived stem cell (ASC)-released exosomes (ASCexos) have multiple biological activities. We examined the effect of ASCexos derived from the inguinal adipose tissue of exercise-trained rats (EX-ASCexos) on adipogenic conversion of 3T3-L1 cells and analyzed their microRNA (miRNA) expression profiles. Differentiation of 3T3-L1 cells into adipocytes was performed for 9 d with EX-ASCexos or ASCexos from sedentary control rats (SED-ASCexos), and the expression of proteins and miRNA involved in adipogenic differentiation were determined. EX-ASCexos but not SED-ASCexos attenuated 3T3-L1 adipocyte differentiation with increased phosph-Ser112PPARγ expression, the inactive form of PPARγ. These differentiated adipocytes were also accompanied by increased phosph-Thr202/Tyr204ERK and decreased dual-specificity phosphatase 3 (DUSP3) levels. The exosomal miRNAs miR-323-5p, miR-433-3p, and miR-874-3p were identified specifically in EX-ASCexos. Of these, miR-323-5p mimic replicated the EX-ASCexo-induced suppression of 3T3-L1 adipocyte differentiation and altered adipogenesis-related factor expression. In conclusion, exercise training-driven exosomal miR-323-5p suppressed 3T3-L1 adipogenesis by increasing phosph-Ser112PPARγ expression, while phosph-Thr202/Tyr204ERK accumulation inhibited DUSP3 expression.
Collapse
Affiliation(s)
- Seita Osawa
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto, 610-0394, Japan; Japan Society for the Promotion of Sci., Tokyo, Japan
| | - Hisashi Kato
- Organization for Research Initiatives and Development, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto, 610-0394, Japan
| | - Daigo Kemmoku
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto, 610-0394, Japan
| | - Sachiko Yamaguchi
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto, 610-0394, Japan
| | - Lureien Jiang
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto, 610-0394, Japan
| | - Yoshifumi Tsuchiya
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto, 610-0394, Japan
| | - Hisashi Takakura
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto, 610-0394, Japan
| | - Tetsuya Izawa
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto, 610-0394, Japan.
| |
Collapse
|
5
|
Guo G, Wang W, Tu M, Zhao B, Han J, Li J, Pan Y, Zhou J, Ma W, Liu Y, Sun T, Han X, An Y. Deciphering adipose development: Function, differentiation and regulation. Dev Dyn 2024; 253:956-997. [PMID: 38516819 DOI: 10.1002/dvdy.708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024] Open
Abstract
The overdevelopment of adipose tissues, accompanied by excess lipid accumulation and energy storage, leads to adipose deposition and obesity. With the increasing incidence of obesity in recent years, obesity is becoming a major risk factor for human health, causing various relevant diseases (including hypertension, diabetes, osteoarthritis and cancers). Therefore, it is of significance to antagonize obesity to reduce the risk of obesity-related diseases. Excess lipid accumulation in adipose tissues is mediated by adipocyte hypertrophy (expansion of pre-existing adipocytes) or hyperplasia (increase of newly-formed adipocytes). It is necessary to prevent excessive accumulation of adipose tissues by controlling adipose development. Adipogenesis is exquisitely regulated by many factors in vivo and in vitro, including hormones, cytokines, gender and dietary components. The present review has concluded a comprehensive understanding of adipose development including its origin, classification, distribution, function, differentiation and molecular mechanisms underlying adipogenesis, which may provide potential therapeutic strategies for harnessing obesity without impairing adipose tissue function.
Collapse
Affiliation(s)
- Ge Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wanli Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wen Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
6
|
Neha, Chaudhary S, Tiwari P, Parvez S. Amelioration of Phytanic Acid-Induced Neurotoxicity by Nutraceuticals: Mechanistic Insights. Mol Neurobiol 2024; 61:7303-7318. [PMID: 38374317 DOI: 10.1007/s12035-024-03985-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024]
Abstract
Phytanic acid (PA) (3,7,11,15-tetramethylhexadecanoic acid) is a methyl-branched fatty acid that enters the body through food consumption, primarily through red meat, dairy products, and fatty marine foods. The metabolic byproduct of phytol is PA, which is then oxidized by the ruminal microbiota and some marine species. The first methyl group at the 3-position prevents the β-oxidation of branched-chain fatty acid (BCFA). Instead, α-oxidation of PA results in the production of pristanic acid (2,10,14-tetramethylpentadecanoic acid) with CO2. This fatty acid (FA) builds up in individuals with certain peroxisomal disorders and is historically linked to neurological impairment. It also causes oxidative stress in synaptosomes, as demonstrated by an increase in the production of reactive oxygen species (ROS), which is a sign of oxidative stress. This review concludes that the nutraceuticals (melatonin, piperine, quercetin, curcumin, resveratrol, epigallocatechin-3-gallate (EGCG), coenzyme Q10, ω-3 FA) can reduce oxidative stress and enhanced the activity of mitochondria. Furthermore, the use of nutraceuticals completely reversed the neurotoxic effects of PA on NO level and membrane potential. Additionally, the review further emphasizes the urgent need for more research into dairy-derived BCFAs and their impact on human health.
Collapse
Affiliation(s)
- Neha
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110 062, India
| | - Shaista Chaudhary
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110 062, India
| | - Prachi Tiwari
- Department of Physiotherapy, School of Nursing Sciences and Allied Health, Jamia Hamdard, New Delhi, 110 062, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110 062, India.
| |
Collapse
|
7
|
Chen SJ, Chien HC, Tsai SH, Jheng YS, Chen Y, Hsieh PS, Tsui PF, Chien S, Tsai MC. Melatonin Ameliorates Atherosclerotic Plaque Vulnerability by Regulating PPARδ-Associated Smooth Muscle Cell Phenotypic Switching. J Pineal Res 2024; 76:e12988. [PMID: 38982751 DOI: 10.1111/jpi.12988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024]
Abstract
Vulnerable atherosclerotic plaque rupture, the leading cause of fatal atherothrombotic events, is associated with an increased risk of mortality worldwide. Peroxisome proliferator-activated receptor delta (PPARδ) has been shown to modulate vascular smooth muscle cell (SMC) phenotypic switching, and, hence, atherosclerotic plaque stability. Melatonin reportedly plays a beneficial role in cardiovascular diseases; however, the mechanisms underlying improvements in atherosclerotic plaque vulnerability remain unknown. In this study, we assessed the role of melatonin in regulating SMC phenotypic switching and its consequential contribution to the amelioration of atherosclerotic plaque vulnerability and explored the mechanisms underlying this process. We analyzed features of atherosclerotic plaque vulnerability and markers of SMC phenotypic transition in high-cholesterol diet (HCD)-fed apolipoprotein E knockout (ApoE-/-) mice and human aortic SMCs (HASMCs). Melatonin reduced atherosclerotic plaque size and necrotic core area while enhancing collagen content, fibrous cap thickness, and smooth muscle alpha-actin positive cell coverage on the plaque cap, which are all known phenotypic characteristics of vulnerable plaques. In atherosclerotic lesions, melatonin significantly decreased the synthetic SMC phenotype and KLF4 expression and increased the expression of PPARδ, but not PPARα and PPARγ, in HCD-fed ApoE-/- mice. These results were subsequently confirmed in the melatonin-treated HASMCs. Further analysis using PPARδ silencing and immunoprecipitation assays revealed that PPARδ plays a role in the melatonin-induced SMC phenotype switching from synthetic to contractile. Collectively, we provided the first evidence that melatonin mediates its protective effect against plaque destabilization by enhancing PPARδ-mediated SMC phenotypic switching, thereby indicating the potential of melatonin in treating atherosclerosis.
Collapse
MESH Headings
- Animals
- Melatonin/pharmacology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Kruppel-Like Factor 4/metabolism
- Humans
- PPAR delta/metabolism
- PPAR delta/genetics
- Mice, Knockout
- Male
- Mice, Knockout, ApoE
- Phenotype
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Apolipoproteins E/deficiency
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Che Chien
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Sin Jheng
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Yi Chen
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Po-Shiuan Hsieh
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan
| | - Pi-Fen Tsui
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Shu Chien
- Department of Bioengineering and Medicine, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, USA
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
8
|
Nagagata BA, Brito G, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. Melatonin supplementation in obese mothers reduces hypothalamic inflammation and enhances thermogenesis in mice progeny. J Nutr Biochem 2024; 128:109625. [PMID: 38521130 DOI: 10.1016/j.jnutbio.2024.109625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
Maternal obesity might induce obesity and metabolic alterations in the progeny. The study aimed to determine the effect of supplementing obese mothers with Mel (Mel) on thermogenesis and inflammation. C57BL/6 female mice (mothers) were fed from weaning to 12 weeks control diet (C, 17% kJ as fat) or a high-fat diet (HF, 49% kJ as fat) and then matted with male mice fed the control diet. Melatonin (10 mg/kg daily) was supplemented to mothers during gestation and lactation, forming the groups C, CMel, HF, and HFMel (n = 10/group). Twelve-week male offspring were studied (plasma biochemistry, immunohistochemistry, protein, and gene expressions at the hypothalamus - Hyp, subcutaneous white adipose tissue - sWAT, and interscapular brown adipose tissue - iBAT). Comparing HFMel vs. HF offspring, fat deposits and plasmatic proinflammatory markers decreased. Also, HFMel showed decreased Hyp proinflammatory markers and neuropeptide Y (anabolic) expression but improved proopiomelanocortin (catabolic) expression. Besides, HFMel sWAT adipocytes changed to a beige phenotype with-beta-3 adrenergic receptor and uncoupling protein-1 activation, concomitant with browning genes activation, triggering the iBAT thermogenic activity. In conclusion, compelling evidence indicated the beneficial effects of supplementing obese mothers with Mel on the health of their mature male offspring. Mel led to sWAT browning-related gene enhancement, increased iBAT thermogenis, and mitigated hypothalamic inflammation. Also, principal component analysis of the data significantly separated the untreated obese mother progeny from the progeny of treated obese mothers. If confirmed in humans, the findings encourage a future guideline recommending Mel supplementation during pregnancy and breastfeeding.
Collapse
Affiliation(s)
- Brenda A Nagagata
- Metabolism section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Nutrition section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabrielle Brito
- Metabolism section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Pan HC, Yang CN, Lee WJ, Sheehan J, Wu SM, Chen HS, Lin MH, Shen LW, Lee SH, Shen CC, Pan LY, Liu SH, Sheu ML. Melatonin Enhanced Microglia M2 Polarization in Rat Model of Neuro-inflammation Via Regulating ER Stress/PPARδ/SIRT1 Signaling Axis. J Neuroimmune Pharmacol 2024; 19:11. [PMID: 38530514 DOI: 10.1007/s11481-024-10108-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/15/2024] [Indexed: 03/28/2024]
Abstract
Neuro-inflammation involves distinct alterations of microglial phenotypes, containing nocuous pro-inflammatory M1-phenotype and neuroprotective anti-inflammatory M-phenotype. Currently, there is no effective treatment for modulating such alterations. M1/M2 marker of primary microglia influenced by Melatonin were detected via qPCR. Functional activities were explored by western blotting, luciferase activity, EMSA, and ChIP assay. Structure interaction was assessed by molecular docking and LIGPLOT analysis. ER-stress detection was examined by ultrastructure TEM, calapin activity, and ERSE assay. The functional neurobehavioral evaluations were used for investigation of Melatonin on the neuroinflammation in vivo. Melatonin had targeted on Peroxisome Proliferator Activated Receptor Delta (PPARδ) activity, boosted LPS-stimulated alterations in polarization from the M1 to the M2 phenotype, and thereby inhibited NFκB-IKKβ activation in primary microglia. The PPARδ agonist L-165,041 or over-expression of PPARδ plasmid (ov-PPARδ) showed similar results. Molecular docking screening, dynamic simulation approaches, and biological studies of Melatonin showed that the activated site was located at PPARδ (phospho-Thr256-PPARδ). Activated microglia had lowered PPARδ activity as well as the downstream SIRT1 formation via enhancing ER-stress. Melatonin, PPARδ agonist and ov-PPARδ all effectively reversed the above-mentioned effects. Melatonin blocked ER-stress by regulating calapin activity and expression in LPS-activated microglia. Additionally, Melatonin or L-165,041 ameliorated the neurobehavioral deficits in LPS-aggravated neuroinflammatory mice through blocking microglia activities, and also promoted phenotype changes to M2-predominant microglia. Melatonin suppressed neuro-inflammation in vitro and in vivo by tuning microglial activation through the ER-stress-dependent PPARδ/SIRT1 signaling cascade. This treatment strategy is an encouraging pharmacological approach for the remedy of neuro-inflammation associated disorders.
Collapse
Affiliation(s)
- Hung-Chuan Pan
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Doctoral Program in Biotechnology Industrial Management and Innovation, National Chung Hsing University, Taichung, Taiwan
- College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| | - Sheng-Mao Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Hong-Shiu Chen
- Department of Neurosurgery, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Mao-Hsun Lin
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Li-Wei Shen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Hua Lee
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chin-Chang Shen
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Liang-Yi Pan
- School of Medicine, Kaohsiung Medical University, Taichung, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Meei-Ling Sheu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Sciences, College of Life Sciences, National Chung Hsing University, 250, Kuo Kuang Road, Taichung, 402, Taiwan.
| |
Collapse
|
10
|
Lei X, Xu Z, Huang L, Huang Y, Tu S, Xu L, Liu D. The potential influence of melatonin on mitochondrial quality control: a review. Front Pharmacol 2024; 14:1332567. [PMID: 38273825 PMCID: PMC10808166 DOI: 10.3389/fphar.2023.1332567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/31/2023] [Indexed: 01/27/2024] Open
Abstract
Mitochondria are critical for cellular energetic metabolism, intracellular signaling orchestration and programmed death regulation. Therefore, mitochondrial dysfunction is associated with various pathogeneses. The maintenance of mitochondrial homeostasis and functional recovery after injury are coordinated by mitochondrial biogenesis, dynamics and autophagy, which are collectively referred to as mitochondrial quality control. There is increasing evidence that mitochondria are important targets for melatonin to exert protective effects under pathological conditions. Melatonin, an evolutionarily conserved tryptophan metabolite, can be synthesized, transported and metabolized in mitochondria. In this review, we summarize the important role of melatonin in the damaged mitochondria elimination and mitochondrial energy supply recovery by regulating mitochondrial quality control, which may provide new strategies for clinical treatment of mitochondria-related diseases.
Collapse
Affiliation(s)
- Xudan Lei
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Experimental Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenni Xu
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Experimental Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Lingxiao Huang
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Experimental Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yujun Huang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyu Tu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Xu
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Experimental Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Dengqun Liu
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Experimental Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Engin AB, Engin A. Tryptophan Metabolism in Obesity: The Indoleamine 2,3-Dioxygenase-1 Activity and Therapeutic Options. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:629-655. [PMID: 39287867 DOI: 10.1007/978-3-031-63657-8_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obesity activates both innate and adaptive immune responses in adipose tissue. Adipose tissue macrophages are functional antigen-presenting cells that promote the proliferation of interferon-gamma (IFN-γ)-producing cluster of differentiation (CD)4+ T cells in adipose tissue of obese subjects. The increased formation of neopterin and degradation of tryptophan may result in decreased T-cell responsiveness and lead to immunodeficiency. The activity of inducible indoleamine 2,3-dioxygenase-1 (IDO1) plays a major role in pro-inflammatory, IFN-γ-dominated settings. The expression of several kynurenine pathway enzyme genes is significantly increased in obesity. IDO1 in obesity shifts tryptophan metabolism from serotonin and melatonin synthesis to the formation of kynurenines and increases the ratio of kynurenine to tryptophan as well as with neopterin production. Reduction in serotonin (5-hydroxytryptamine; 5-HT) production provokes satiety dysregulation that leads to increased caloric uptake and obesity. According to the monoamine-deficiency hypothesis, a deficiency of cerebral serotonin is involved in neuropsychiatric symptomatology of depression, mania, and psychosis. Indeed, bipolar disorder (BD) and related cognitive deficits are accompanied by a higher prevalence of overweight and obesity. Furthermore, the accumulation of amyloid-β in Alzheimer's disease brains has several toxic effects as well as IDO induction. Hence, abdominal obesity is associated with vascular endothelial dysfunction. kynurenines and their ratios are prognostic parameters in coronary artery disease. Increased kynurenine/tryptophan ratio correlates with increased intima-media thickness and represents advanced atherosclerosis. However, after bariatric surgery, weight reduction does not lead to the normalization of IDO1 activity and atherosclerosis. IDO1 is involved in the mechanisms of immune tolerance and in the concept of tumor immuno-editing process in cancer development. Serum IDO1 activity is still used as a parameter in cancer development and growth. IDO-producing tumors show a high total IDO immunostaining score, and thus, using IDO inhibitors, such as Epacadostat, Navoximod, and L isomer of 1-methyl-tryptophan, seems an important modality for cancer treatment. There is an inverse correlation between serum folate concentration and body mass index, thus folate deficiency leads to hyperhomocysteinemia-induced oxidative stress. Immune checkpoint blockade targeting cytotoxic T-lymphocyte-associated protein-4 synergizes with imatinib, which is an inhibitor of mitochondrial folate-mediated one-carbon (1C) metabolism. Antitumor effects of imatinib are enhanced by increasing T-cell effector function in the presence of IDO inhibition. Combining IDO targeting with chemotherapy, radiotherapy and/or immunotherapy, may be an effective tool against a wide range of malignancies. However, there are some controversial results regarding the efficacy of IDO1 inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
12
|
Guo C, He J, Deng X, Wang D, Yuan G. Potential therapeutic value of melatonin in diabetic nephropathy: improvement beyond anti-oxidative stress. Arch Physiol Biochem 2023; 129:1250-1261. [PMID: 34048666 DOI: 10.1080/13813455.2021.1933539] [Citation(s) in RCA: 5] [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: 04/11/2021] [Accepted: 05/18/2021] [Indexed: 12/23/2022]
Abstract
Diabetic nephropathy (DN) is a common complication of diabetes, and it is also the main cause of chronic renal failure. Physiological/pathological changes mediated by high glucose are the main factors causing injury of DN, including the enhancement of polyol pathway, the accumulation of advanced glycation products (AGEs), and the activation of protein kinase C (PKC) and transforming growth factor-β (TGF-β) signals. In addition, the abnormal activation of renin-angiotensin system (RAS) and oxidative stress are also involved. Melatonin is a physiological hormone mainly secreted by the pineal gland which has been proved to be related to diabetes. Studies have shown that exogenous melatonin intervention can reduce blood glucose and alleviate high glucose mediated pathological damage. At the same time, melatonin also has a strong antioxidant effect, and can inhibit the activation of RAS. Therefore, it is of great significance to explore the therapeutic effect and value of melatonin on DN.
Collapse
Affiliation(s)
- Chang Guo
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jianqiang He
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xia Deng
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Dong Wang
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guoyue Yuan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
13
|
Huang Y, Liang B, Li Z, Zhong Y, Wang B, Zhang B, Du J, Ye R, Xian H, Min W, Yan X, Deng Y, Feng Y, Bai R, Fan B, Yang X, Huang Z. Polystyrene nanoplastic exposure induces excessive mitophagy by activating AMPK/ULK1 pathway in differentiated SH-SY5Y cells and dopaminergic neurons in vivo. Part Fibre Toxicol 2023; 20:44. [PMID: 37993864 PMCID: PMC10664492 DOI: 10.1186/s12989-023-00556-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Microplastics and nanoplastics (MNPs) are emerging environmental contaminants detected in human samples, and have raised concerns regarding their potential risks to human health, particularly neurotoxicity. This study aimed to investigate the deleterious effects of polystyrene nanoplastics (PS-NPs, 50 nm) and understand their mechanisms in inducing Parkinson's disease (PD)-like neurodegeneration, along with exploring preventive strategies. METHODS Following exposure to PS-NPs (0.5-500 μg/mL), we assessed cytotoxicity, mitochondrial integrity, ATP levels, and mitochondrial respiration in dopaminergic-differentiated SH-SY5Y cells. Molecular docking and dynamic simulations explored PS-NPs' interactions with mitochondrial complexes. We further probed mitophagy's pivotal role in PS-NP-induced mitochondrial damage and examined melatonin's ameliorative potential in vitro. We validated melatonin's intervention (intraperitoneal, 10 mg/kg/d) in C57BL/6 J mice exposed to 250 mg/kg/d of PS-NPs for 28 days. RESULTS In our in vitro experiments, we observed PS-NP accumulation in cells, including mitochondria, leading to cell toxicity and reduced viability. Notably, antioxidant treatment failed to fully rescue viability, suggesting reactive oxygen species (ROS)-independent cytotoxicity. PS-NPs caused significant mitochondrial damage, characterized by altered morphology, reduced mitochondrial membrane potential, and decreased ATP production. Subsequent investigations pointed to PS-NP-induced disruption of mitochondrial respiration, potentially through interference with complex I (CI), a concept supported by molecular docking studies highlighting the influence of PS-NPs on CI. Rescue experiments using an AMPK pathway inhibitor (compound C) and an autophagy inhibitor (3-methyladenine) revealed that excessive mitophagy was induced through AMPK/ULK1 pathway activation, worsening mitochondrial damage and subsequent cell death in differentiated SH-SY5Y cells. Notably, we identified melatonin as a potential protective agent, capable of alleviating PS-NP-induced mitochondrial dysfunction. Lastly, our in vivo experiments demonstrated that melatonin could mitigate dopaminergic neuron loss and motor impairments by restoring mitophagy regulation in mice. CONCLUSIONS Our study demonstrated that PS-NPs disrupt mitochondrial function by affecting CI, leading to excessive mitophagy through the AMPK/ULK1 pathway, causing dopaminergic neuron death. Melatonin can counteract PS-NP-induced mitochondrial dysfunction and motor impairments by regulating mitochondrial autophagy. These findings offer novel insights into the MNP-induced PD-like neurodegenerative mechanisms, and highlight melatonin's protective potential in mitigating the MNP's environmental risk.
Collapse
Affiliation(s)
- Yuji Huang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Boxuan Liang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zhiming Li
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yizhou Zhong
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Bo Wang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Bingli Zhang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jiaxin Du
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Rongyi Ye
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Hongyi Xian
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Weicui Min
- School of Environmental Science and Engineering, Shandong University, Qingdao, 266237, People's Republic of China
| | - Xiliang Yan
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, People's Republic of China
| | - Yanhong Deng
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yu Feng
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ruobing Bai
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Bingchi Fan
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xingfen Yang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Food Safety and Health Research Center, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zhenlie Huang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
14
|
Gao X, Sun H, Hao S, Sun H, Ge J. Melatonin protects HT-22 cells against palmitic acid-induced glucolipid metabolic dysfunction and cell injuries: Involved in the regulation of synaptic plasticity and circadian rhythms. Biochem Pharmacol 2023; 217:115846. [PMID: 37804870 DOI: 10.1016/j.bcp.2023.115846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Melatonin (MLT) is ahormonal substance reported with various pharmacological activities.Based on its effects of neuroprotection and metabolic regulation, the aim of the present study is to investigate its potential effect on palmitic acid (PA)-induced cell injuries and glucolipid metabolic dysfunction and explore the possible mechanism. Briefly, HT-22 cells were challenged with PA (0.1 mM, 24 h) and treated with MLT (10-6-10-8 mol/L). Cell proliferation, lipid accumulation and glucose consumption were detected. The protein expression of key molecular involved with the function of synaptic plasticity and circadian rhythms were measured via western blotting, and the expression of Map-2, MT1A, MT1B and Bmal1 were measured via immunofluorescence staining. The results showed that MLT could alleviate the neurotoxicity induced by PA, as indicated by the increased cell proliferation, enhanced fluorescence intensity of Map-2, and decreased lipid deposition and insulin resistance. Moreover, treatment of MLT could reverse the imbalanced expression of p-Akt, p-ERK, Synapsin I, Synaptotagmin I, BDNF, MT1B, Bmal1, and Clock in PA-induced HT-22 cells. These results suggested a remarkably neuroprotective effect of MLT against PA-induced cell injury and glucolipid metabolic dysfunction, the mechanism of which might be involved in the regulation of synaptic plasticity and circadian rhythms.
Collapse
Affiliation(s)
- Xinran Gao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Huaizhi Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Shengwei Hao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Huimin Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Jinfang Ge
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China.
| |
Collapse
|
15
|
Esteban-Zubero E, López-Pingarrón L, Ramírez JM, Reyes-Gonzales MC, Azúa-Romeo FJ, Soria-Aznar M, Agil A, García JJ. Melatonin Preserves Fluidity in Cell and Mitochondrial Membranes against Hepatic Ischemia-Reperfusion. Biomedicines 2023; 11:1940. [PMID: 37509579 PMCID: PMC10377318 DOI: 10.3390/biomedicines11071940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
We evaluated the in vivo effects of melatonin treatment on oxidative damage in the liver in an experimental model of ischemia-reperfusion. A total of 37 male Sprague-Dawley rats were randomly divided into four groups: control, ischemia, ischemia + reperfusion, and ischemia + reperfusion + melatonin. Hepatic ischemia was maintained for 20 min, and the clamp was removed to initiate vascular reperfusion for 30 min. Melatonin (50 mg/kg body weight) was intraperitoneally administered. Fluidity was measured by polarization changes in 1-(4-trimethylammoniumphenyl)-6-phenyl-1,3,5-hexatriene-p-toluene sulfonate). After 20 min of ischemia, no significant changes were observed in cell and mitochondrial membrane fluidity levels, lipid peroxidation, and protein carbonylation. However, after 30 min of reperfusion, membrane fluidity decreased compared to controls. Increases in lipid and protein oxidation were also seen in hepatic homogenates of animals exposed to reperfusion. Melatonin injected 30 min before ischemia and reperfusion fully prevented membrane rigidity and both lipid and protein oxidation. Livers from ischemia-reperfusion showed histopathological alterations and positive labeling with antibodies to oxidized lipids and proteins. Melatonin reduced the severity of these morphological changes and protected against in vivo ischemia-reperfusion-induced toxicity in the liver. Therefore, melatonin might be a candidate for co-treatment for patients with hepatic vascular occlusion followed by reperfusion.
Collapse
Affiliation(s)
- Eduardo Esteban-Zubero
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Laura López-Pingarrón
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | | | - Marcos César Reyes-Gonzales
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Francisco Javier Azúa-Romeo
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Marisol Soria-Aznar
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Ahmad Agil
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - José Joaquín García
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
16
|
Lee S, Kim HJ, Cho HB, Kim HR, Lee S, Park JI, Park KH. Melatonin loaded PLGA nanoparticles effectively ameliorate the in vitro maturation of deteriorated oocytes and the cryoprotective abilities during vitrification process. Biomater Sci 2023; 11:2912-2923. [PMID: 36883517 DOI: 10.1039/d2bm02054h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Almost all cells can be exposed to stress, but oocytes, which are female germ cells, are particularly vulnerable to damage. In this study, melatonin, a well-known antioxidant, was loaded into biodegradable poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) and delivered to damaged oocytes in order to improve their quality and restoration. Etoposide (ETP)-induced deteriorated oocytes show poor maturity, mitochondrial aggregation, and DNA damage. Treatment of NPs not only reduced DNA damage but also improved mitochondrial stability, as evidenced by increased ATP levels and mitochondrial homogeneity. When melatonin was added to the culture medium at the same concentration as that present in NPs, DNA and mitochondrial repair was insignificant due to the half-life of melatonin, whereas DNA repair in damaged oocytes upon multiple treatments with melatonin was similar to that observed with melatonin-loaded NPs. Next, we evaluated whether the oocytes treated with NPs could have cryoprotective abilities during vitrification/thawing. Vitrified-oocytes were stored at -196 °C for 0.25 h (T1) or 0.5 h (T2). After thawing, live oocytes were subjected to in vitro maturation. The NP-treated group showed maturity similar to the control group (77.8% in T1, 72.7% in T2) and the degree of DNA damage was reduced compared to the ETP-induced group (p < 0.05).
Collapse
Affiliation(s)
- Sujin Lee
- Department of Biomedical Science, College of Life Science, CHA University, 6F, CHA Bio-Complex, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 134-88, Republic of Kore.
| | - Hye Jin Kim
- Department of Biomedical Science, College of Life Science, CHA University, 6F, CHA Bio-Complex, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 134-88, Republic of Kore.
| | - Hui Bang Cho
- Department of Biomedical Science, College of Life Science, CHA University, 6F, CHA Bio-Complex, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 134-88, Republic of Kore.
| | - Hye-Ryoung Kim
- Department of Biomedical Science, College of Life Science, CHA University, 6F, CHA Bio-Complex, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 134-88, Republic of Kore.
| | - Sujeong Lee
- Department of Biomedical Science, College of Life Science, CHA University, 6F, CHA Bio-Complex, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 134-88, Republic of Kore.
| | - Ji-In Park
- Department of Biomedical Science, College of Life Science, CHA University, 6F, CHA Bio-Complex, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 134-88, Republic of Kore.
| | - Keun-Hong Park
- Department of Biomedical Science, College of Life Science, CHA University, 6F, CHA Bio-Complex, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 134-88, Republic of Kore.
| |
Collapse
|
17
|
Li J, Huang T, Xia M, Lu J, Xu X, Liu H, Zhang W. Exogenous melatonin mediates radish ( Raphanus sativus) and Alternaria brassicae interaction in a dose-dependent manner. FRONTIERS IN PLANT SCIENCE 2023; 14:1126669. [PMID: 36923135 PMCID: PMC10009256 DOI: 10.3389/fpls.2023.1126669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Radish (Raphanus sativus L.) is an economically important vegetable worldwide, but its sustainable production and breeding are highly threatened by blight disease caused by Alternaria brassicae. Melatonin is an important growth regulator that can influence physiological activities in both plants and microbes and stimulate biotic stress resistance in plants. In this study, 0-1500 μM melatonin was exogenously applied to healthy radish seedlings, in vitro incubated A. brassicae, and diseased radish seedlings to determine the effects of melatonin on host, pathogen, and host-pathogen interaction. At sufficient concentrations (0-500 μM), melatonin enhanced growth and immunity of healthy radish seedlings by improving the function of organelles and promoting the biosynthesis of antioxidant enzymes, chitin, organic acid, and defense proteins. Interestingly, melatonin also improved colony growth, development, and virulence of A. brassicae. A strong dosage-dependent effect of melatonin was observed: 50-500 μM promoted host and pathogen vitality and resistance (500 μM was optimal) and 1500 μM inhibited these processes. Significantly less blight was observed on diseased seedlings treated with 500 μM melatonin, indicating that melatonin more strongly enhanced the growth and immunity of radish than it promoted the development and virulence of A. brassicae at this treatment concentration. These effects of MT were mediated by transcriptional changes of key genes as identified by RNA-seq, Dual RNA-seq, and qRT-PCR. The results from this work provide a theoretical basis for the application of melatonin to protect vegetable crops against pathogens.
Collapse
Affiliation(s)
- Jingwei Li
- Institute of Vegetable Industry Technology Research, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Tingmin Huang
- Institute of Vegetable Industry Technology Research, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Ming Xia
- Institute of Vegetable Industry Technology Research, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
- School of Computing, Chongqing College of Humanities, Science and Technology, Hechuan, China
| | - Jinbiao Lu
- Institute of Vegetable Industry Technology Research, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Xiuhong Xu
- Institute of Vegetable Industry Technology Research, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Haiyi Liu
- Institute of Vegetable Industry Technology Research, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Wanping Zhang
- Institute of Vegetable Industry Technology Research, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| |
Collapse
|
18
|
Yuan Q, Zeng ZL, Yang S, Li A, Zu X, Liu J. Mitochondrial Stress in Metabolic Inflammation: Modest Benefits and Full Losses. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8803404. [PMID: 36457729 PMCID: PMC9708372 DOI: 10.1155/2022/8803404] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 09/02/2023]
Abstract
Energy intake and metabolic balance are the pillars of health preservation. Overnutrition causes nonspecific, persistently low inflammatory state known as metabolic inflammation. This condition contributes to the pathophysiology of various metabolic disorders, such as atherosclerosis, obesity, diabetes mellitus, and metabolic syndrome. The mitochondria maintain the balance of energy metabolism. Excessive energy stress can lead to mitochondrial dysfunction, which promotes metabolic inflammation. The inflammatory environment further impairs mitochondrial function. Accordingly, excellent organism design keeps the body metabolically healthy in the context of mitochondrial dysfunction, and moderate mitochondrial stress can have a beneficial effect. This review summarises the research progress on the multifaceted characterisation of mitochondrial dysfunction and its role in metabolic inflammation.
Collapse
Affiliation(s)
- Qing Yuan
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Z. L. Zeng
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Shiqi Yang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Anqi Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuyu Zu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
19
|
Huang X, Chen W, Gu C, Liu H, Hou M, Qin W, Zhu X, Chen X, Liu T, Yang H, He F. Melatonin suppresses bone marrow adiposity in ovariectomized rats by rescuing the imbalance between osteogenesis and adipogenesis through SIRT1 activation. J Orthop Translat 2022; 38:84-97. [PMID: 36381247 PMCID: PMC9619141 DOI: 10.1016/j.jot.2022.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/17/2022] [Accepted: 10/06/2022] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Accelerated imbalance between bone formation and bone resorption is associated with bone loss in postmenopausal osteoporosis. Studies have shown that this loss is accompanied by an increase in bone marrow adiposity. Melatonin was shown to improve impaired bone formation capacity of bone marrow-derived mesenchymal stem cells from ovariectomized rats (OVX-BMMSCs). OBJECTIVES To investigate whether the anti-osteoporosis effect of melatonin involves regulation of the equilibrium between osteogenic and adipogenic differentiation of osteoporotic BMMSCs. METHODS To induce osteoporosis, female Sprague-Dawley rats received ovariectomy (OVX). Primary BMMSCs were isolated from tibiae and femurs of OVX and sham-op rats and were induced towards osteogenic or adipogenic differentiation. Matrix mineralization was determined by Alizarin Red S (ARS) and lipid formation was evaluated by Oil Red O. OVX rats were injected with melatonin through the tail vein. Bone microarchitecture was determined using micro computed tomography and marrow adiposity were examined by histology staining. RESULTS OVX-BMMSCs exhibited a compromised osteogenic potential and an enhanced lineage differentiation towards adipocytes. In vitro melatonin improved osteogenic differentiation of OVX-BMMSCs and promoted matrix mineralization by enhancing the expression of transcription factor RUNX2 in a dose-dependent manner. Moreover, melatonin significantly inhibited lipid formation and suppressed OVX-BMMSCs adipogenesis by down-regulating peroxisome proliferator-activated receptor γ (PPARγ). Intravenous injection of melatonin prevented bone mass reduction and bone architecture destruction in ovariectomized rats. Importantly, there was a significant inhibition of adipose tissue formation in the bone marrow. Mechanistic investigations revealed that SIRT1 was involved in melatonin-mediated determination of stem cell fate. Inhibition of SIRT1 abolished the protective effects of melatonin on bone formation by inducing BMMSCs towards adipocyte differentiation. CONCLUSIONS Melatonin reversed the differentiation switch of OVX-BMMSCs from osteogenesis to adipogenesis by activating the SIRT1 signaling pathway. Restoration of stem cell lineage commitment by melatonin prevented marrow adipose tissue over-accumulation and protected from bone loss in postmenopausal osteoporosis. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Determination of stem cell fate towards osteoblasts or adipocytes plays a pivotal role in regulating bone metabolism. This study demonstrates the protective effect of melatonin on bone mass in estrogen-deficient rats by suppressing adipose tissue accumulation in the bone marrow. Melatonin may serve as a promising candidate for the treatment of osteoporosis in clinics.
Collapse
Affiliation(s)
- Xiaoxiong Huang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China,Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), No. 41 Northwest Street, Ningbo, 315010, Zhejiang, China,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Weikai Chen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Chao Gu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Hao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Wanjin Qin
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xi Chen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China,Corresponding author. Department of Pathology, The Third Affiliated Hospital of Soochow University, No.185 Juqian Road, Changzhou, 213003, Jiangsu, China.
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China,Corresponding author. Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China,Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China,Corresponding author. Orthopaedic Institute, Soochow University, Suzhou 215000, China
| |
Collapse
|
20
|
Targhazeh N, Hutt KJ, Winship AL, Reiter R, Yousefi B. Melatonin as an oncostatic agent: Review of the modulation of tumor microenvironment and overcoming multidrug resistance. Biochimie 2022; 202:71-84. [PMID: 36116742 DOI: 10.1016/j.biochi.2022.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022]
Abstract
Multi drug resistance (MDR) generally limits the efficacy of chemotherapy in cancer patients and can be categorized into primary or acquired resistance. Melatonin (MLT), a lipophilic hormone released from pineal gland, is a molecule with oncostatic effects. Here, we will briefly review the contribution of different microenvironmental components including fibroblasts, immune and inflammatory cells, stem cells and vascular endothelial cells in tumor initiation, progression and development. Then, the mechanisms by which MLT can potentially affect these elements and regulate drug resistance will be presented. Finally, we will explain how different studies have used novel strategies incorporating MLT to suppress cancer resistance against therapeutics.
Collapse
Affiliation(s)
- Niloufar Targhazeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karla J Hutt
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Amy L Winship
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Russel Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA.
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Pivonello C, Negri M, Patalano R, Amatrudo F, Montò T, Liccardi A, Graziadio C, Muscogiuri G, Pivonello R, Colao A. The role of melatonin in the molecular mechanisms underlying metaflammation and infections in obesity: A narrative review. Obes Rev 2022; 23:e13390. [PMID: 34861097 PMCID: PMC9285339 DOI: 10.1111/obr.13390] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/20/2022]
Abstract
Obesity is a chronic condition whose management is a critical challenge for physicians. The scientific community has increased its focus on the molecular mechanisms involved in obesity etiopathogenesis to better manage patients with obesity and its associated complications. The tight connection between adipose tissue and the immune system has been demonstrated to play a crucial role in inflammation, and melatonin is important for circadian rhythm regulation and metabolic homeostasis, in which it orchestrates several molecular mechanisms involved in obesity and associated inflammation. Melatonin also regulates innate and adaptive immunity; its antioxidant properties are linked to reduced predisposition to infection and weight gain in patients with obesity through the modulation of the immune response, which has a significant beneficial effect on inflammation and, consequently, on the metabolic state. Low melatonin levels have been linked to obesity, and melatonin supplementation can reduce body weight, improve metabolic profile, and ameliorate immune responses and pro-inflammatory stimuli. The role of melatonin in obesity is mainly related to improved oxidative stress signaling, modulation of adipokine secretion, and a switching from white-to-brown adipose tissue phenotype and activity. Moreover, the role of melatonin in obesity modulation by controlling circadian rhythm has recently emerged as a pivotal mechanism for lipid and glucose metabolism dysfunction in adipose, muscle, and liver tissues. Melatonin may also regulate the immune system by acting directly on thymus morphology and activity as well as by modulating oxidative stress and inflammatory states during infections. The tight association between melatonin and immune response regulation is coordinated by Toll-like receptors, which are rhythmically expressed during the day. Their expression may be strongly modulated by melatonin as their signaling is highly inhibited by melatonin. The current review summarizes studies of melatonin-induced mechanisms involved in infection regulation, particularly the modulation of obesity-associated inflammation and systemic complications.
Collapse
Affiliation(s)
- Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Roberta Patalano
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Feliciana Amatrudo
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Tatiana Montò
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Alessia Liccardi
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Chiara Graziadio
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,UNESCO Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,UNESCO Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Naples, Italy.,UNESCO Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| |
Collapse
|
22
|
Therapeutic potential of melatonin in colorectal cancer: Focus on lipid metabolism and gut microbiota. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166281. [PMID: 34610472 DOI: 10.1016/j.bbadis.2021.166281] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/24/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is one of the most common gastrointestinal malignancies. The occurrence and development of CRC are complicated processes. Obesity and dysbacteriosis have been increasingly regarded as the main risk factors for CRC. Understanding the etiology of CRC from multiple perspectives is conducive to screening for some potential drugs or new treatment strategies to limit the serious side effects of conventional treatment and prolong the survival of CRC patients. Melatonin, a natural indoleamine, is mainly produced by the pineal gland, but it is also abundant in other tissues, including the gastrointestinal tract, retina, testes, lymphocytes, and Harder's glands. Melatonin could participate in lipid metabolism by regulating adipogenesis and lipolysis. Additionally, many studies have focused on the potential beneficial effects of melatonin in CRC, such as promotion of apoptosis; inhibition of cell proliferation, migration, and invasion; antioxidant activity; and immune regulation. Meaningfully, gut microbiota is the main determinant of all aspects of health and disease (including obesity and tumorigenesis). The gut microbiota is of great significance for understanding the relationship between obesity and increased risk of CRC. Although the current understanding of how the melatonin-mediated gut microbiota coordinates a variety of physiological and pathological activities is fairly comprehensive, there are still many unknown topics to be explored in the face of a complex nutritional status and a changeable microbiota. This review summarizes the potential links among melatonin, lipid metabolism, gut microbiota, and CRC to promote the development of melatonin as a preventive and therapeutic agent for CRC.
Collapse
|
23
|
Jin L, Zhu HY, Kang XJ, Lin LP, Zhang PY, Tan T, Yu Y, Fan Y. Melatonin protects against oxybenzone-induced deterioration of mouse oocytes during maturation. Aging (Albany NY) 2020; 13:2727-2749. [PMID: 33373318 PMCID: PMC7880374 DOI: 10.18632/aging.202323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/11/2020] [Indexed: 01/12/2023]
Abstract
Oxybenzone (OBZ), an ultraviolet light filter that is widely used in sunscreens and cosmetics, is an emerging contaminant found in humans and the environment. Recent studies have shown that OBZ has been detected in women's plasma, urine, and breast milk. However, the effects of OBZ exposure on oocyte meiosis have not been addressed. In this study, we investigated the detrimental effects of OBZ on oocyte maturation and the protective roles of melatonin (MT) in OBZ-exposed mouse models. Our in vitro and in vivo results showed that OBZ suppressed oocyte maturation, while MT attenuated the meiotic defects induced by OBZ. In addition, OBZ facilitated H3K4 demethylation by increasing the expression of the Kdm5 family of genes, elevating ROS levels, decreasing GSH, impairing mitochondrial quality, and disrupting spindle configuration in oocytes. However, MT treatment resulted in significant protection against OBZ-induced damage during oocyte maturation and improved oocyte quality. The mechanisms underlying the beneficial roles of MT involved reduction of oxidative stress, inhibition of apoptosis, restoration of abnormal spindle assembly and up-regulation of H3K4me3. Collectively, our results suggest that MT protects against defects induced by OBZ during mouse oocyte maturation in vitro and in vivo.
Collapse
Affiliation(s)
- Long Jin
- Department of Gynecology and Obstetrics, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, Guangdong, China
| | - Hai-Ying Zhu
- Department of Gynecology and Obstetrics, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, Guangdong, China
| | - Xiang-Jin Kang
- Department of Gynecology and Obstetrics, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, Guangdong, China
| | - Li-Ping Lin
- Department of Gynecology and Obstetrics, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, Guangdong, China
| | - Pu-Yao Zhang
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Tao Tan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Scienceand Technology, Kunming 650500, Yunnan, China
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Yong Fan
- Department of Gynecology and Obstetrics, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, Guangdong, China
| |
Collapse
|
24
|
Exosome and Melatonin Additively Attenuates Inflammation by Transferring miR-34a, miR-124, and miR-135b. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1621394. [PMID: 33299858 PMCID: PMC7707940 DOI: 10.1155/2020/1621394] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022]
Abstract
The positive effects of mesenchymal stem cells (MSCs) are primarily activated through molecular secretions known as paracrine activity, which regulates the function of various cell types including immune cells. Accumulating evidence shows that exosomes of soluble factors released from MSCs are potential alternative agents for stem cell-based therapy, although the exact underlying mechanism has not been elucidated. The purpose of this study was to evaluate the potential effects of exosomes produced by adipose-derived MSCs and to examine the changes in anti-inflammatory genes in concurrence with the polarization of M2 macrophages in cellular models ex vivo. Isolated exosomes were used to investigate the inflammatory modulation in pro-inflammatory cytokine-treated fibroblasts and THP-1 cells. The anti-inflammatory mRNA expression associated with M2 macrophages was significantly upregulated after exosome treatment in an interferon gamma and tumor necrosis factor alpha-treated inflammatory environment. Furthermore, melatonin-stimulated exosomes exerted superior anti-inflammatory modulation via exosomal miRNAs miR-34a, miR-124, and miR-135b, compared with exosomes. Our results indicate that melatonin-stimulated exosomes originating from adipose-derived MSCs are safe and efficient tools for regenerative medicine to treat inflammatory diseases.
Collapse
|
25
|
Kato H, Ogasawara J, Takakura H, Shirato K, Sakurai T, Kizaki T, Izawa T. Exercise Training-Enhanced Lipolytic Potency to Catecholamine Depends on the Time of the Day. Int J Mol Sci 2020; 21:ijms21186920. [PMID: 32967199 PMCID: PMC7554872 DOI: 10.3390/ijms21186920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/16/2020] [Accepted: 09/19/2020] [Indexed: 12/22/2022] Open
Abstract
Exercise training is well known to enhance adipocyte lipolysis in response to hormone challenge. However, the existence of a relationship between the timing of exercise training and its effect on adipocyte lipolysis is unknown. To clarify this issue, Wistar rats were run on a treadmill for 9 weeks in either the early part (E-EX) or late part of the active phase (L-EX). L-EX rats exhibited greater isoproterenol-stimulated lipolysis expressed as fold induction over basal lipolysis, with greater protein expression levels of hormone-sensitive lipase (HSL) phosphorylated at Ser 660 compared to E-EX rats. Furthermore, we discovered that Brain and muscle Arnt-like (BMAL)1 protein can associate directly with several protein kinase A (PKA) regulatory units (RIα, RIβ, and RIIβ) of protein kinase, its anchoring protein (AKAP)150, and HSL, and that the association of BMAL1 with the regulatory subunits of PKA, AKAP150, and HSL was greater in L-EX than in E-EX rats. In contrast, comparison between E-EX and their counterpart sedentary control rats showed a greater co-immunoprecipitation only between BMAL1 and ATGL. Thus, both E-EX and L-EX showed an enhanced lipolytic response to isoproterenol, but the mechanisms underlying exercise training-enhanced lipolytic response to isoproterenol were different in each group.
Collapse
Affiliation(s)
- Hisashi Kato
- Organization for Research Initiatives and Development, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan;
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan;
| | - Junetsu Ogasawara
- Department of Health Science, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan;
| | - Hisashi Takakura
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan;
| | - Ken Shirato
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University of School Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan; (K.S.); (T.S.); (T.K.)
| | - Takuya Sakurai
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University of School Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan; (K.S.); (T.S.); (T.K.)
| | - Takako Kizaki
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University of School Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan; (K.S.); (T.S.); (T.K.)
| | - Tetsuya Izawa
- Faculty of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan;
- Graduate School of Health and Sports Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe City, Kyoto 610-0394, Japan
- Correspondence: ; Tel.: +81-424-65-6721; Fax: +81-424-65-6729
| |
Collapse
|
26
|
Hu ZP, Fang XL, Sheng B, Guo Y, Yu YQ. Melatonin inhibits macrophage infiltration and promotes plaque stabilization by upregulating anti-inflammatory HGF/c-Met system in the atherosclerotic rabbit: USPIO-enhanced MRI assessment. Vascul Pharmacol 2020; 127:106659. [DOI: 10.1016/j.vph.2020.106659] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/06/2020] [Accepted: 02/14/2020] [Indexed: 01/08/2023]
|
27
|
Park JH, Seo I, Shim HM, Cho H. Melatonin ameliorates SGLT2 inhibitor-induced diabetic ketoacidosis by inhibiting lipolysis and hepatic ketogenesis in type 2 diabetic mice. J Pineal Res 2020; 68:e12623. [PMID: 31743484 DOI: 10.1111/jpi.12623] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022]
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are effective hypoglycemic agents that can induce glycosuria. However, there are increasing concerns that they might induce diabetic ketoacidosis. This study investigated the effect of melatonin on SGTL2i-induced ketoacidosis in insulin-deficient type 2 diabetic (T2D) mice. The SGLT2i dapagliflozin reduced blood glucose level and plasma insulin concentrations in T2D mice, but induced increases in the concentrations of plasma β-hydroxybutyrate, acetoacetate, and free fatty acid and a decrease in the concentration of plasma bicarbonate, resulting in ketoacidosis. Melatonin inhibited dapagliflozin-induced ketoacidosis without inducing any change in blood glucose level or plasma insulin concentration. In white adipose tissue, melatonin inhibited lipolysis and downregulated phosphorylation of PKA, HSL, and perilipin-1. In liver tissue, melatonin suppressed cellular cyclic AMP levels and downregulated phosphorylation of PKA, AMPK, and acetyl-CoA carboxylase (ACC). In addition, melatonin increased hepatic ACC activity, but decreased hepatic CPT1a activity and acetyl-CoA content. These effects of melatonin on lipolysis and hepatic ketogenesis were blocked by pretreatment with melatonin receptor antagonist or PKA activator. Collectively, these results suggest that melatonin can ameliorate SGLT2i-induced ketoacidosis by inhibiting lipolysis and hepatic ketogenesis though cyclic AMP/PKA signaling pathways in T2D mice. Thus, melatonin treatment may offer protection against SGLT2i-induced ketoacidosis.
Collapse
Affiliation(s)
- Jae-Hyung Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Incheol Seo
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Hae-Min Shim
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Hochan Cho
- Division of Endocrinology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
28
|
Xu Z, You W, Liu J, Wang Y, Shan T. Elucidating the Regulatory Role of Melatonin in Brown, White, and Beige Adipocytes. Adv Nutr 2020; 11:447-460. [PMID: 31355852 PMCID: PMC7442421 DOI: 10.1093/advances/nmz070] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/28/2019] [Accepted: 06/13/2019] [Indexed: 12/15/2022] Open
Abstract
The high prevalence of obesity and its associated metabolic diseases has heightened the importance of understanding control of adipose tissue development and energy metabolism. In mammals, 3 types of adipocytes with different characteristics and origins have been identified: white, brown, and beige. Beige and brown adipocytes contain numerous mitochondria and have the capability to burn energy and counteract obesity, while white adipocytes store energy and are closely associated with metabolic disorders and obesity. Thus, regulation of the development and function of different adipocytes is important for controlling energy balance and combating obesity and related metabolic disorders. Melatonin is a neurohormone, which plays multiple roles in regulating inflammation, blood pressure, insulin actions, and energy metabolism. This article summarizes and discusses the role of melatonin in white, beige, and brown adipocytes, especially in affecting adipogenesis, inducing beige formation or white adipose tissue browning, enhancing brown adipose tissue mass and activities, improving anti-inflammatory and antioxidative effects, regulating adipokine secretion, and preventing body weight gain. Based on the current findings, melatonin is a potential therapeutic agent to control energy metabolism, adipogenesis, fat deposition, adiposity, and related metabolic diseases.
Collapse
Affiliation(s)
- Ziye Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China,Address correspondence to TS (E-mail: )
| |
Collapse
|
29
|
Circadian rhythms, Neuroinflammation and Oxidative Stress in the Story of Parkinson's Disease. Cells 2020; 9:cells9020314. [PMID: 32012898 PMCID: PMC7072287 DOI: 10.3390/cells9020314] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is one of the main neurodegenerative disease characterized by a progressive degeneration of neurons constituted by dopamine in the substantia nigra pars compacta. The etiologies of PD remain unclear. Aging is the main risk factor for PD. Aging could dysregulate molecular pathways controlling cell homeostatic mechanisms. PD cells are the sites of several metabolic abnormalities including neuroinflammation and oxidative stress. Metabolic structures are driven by circadian rhythms. Biologic rhythms are complex systems interacting with the environment and controlling several physiological pathways. Recent findings have shown that the dysregulation of the circadian rhythms is correlated with PD and its metabolic dysregulations. This review is focused on the key role of circadian rhythms and their impact on neuroinflammation and oxidative stress in Parkinson’s disease.
Collapse
|
30
|
Heo JS, Pyo S, Lim JY, Yoon DW, Kim BY, Kim JH, Kim GJ, Lee SG, Kim J. Biological effects of melatonin on human adipose‑derived mesenchymal stem cells. Int J Mol Med 2019; 44:2234-2244. [PMID: 31573052 PMCID: PMC6844604 DOI: 10.3892/ijmm.2019.4356] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into other cell types and exhibit immunomodulatory effects. MSCs are affected by several intrinsic and extrinsic signaling modulators, including growth factors, cytokines, extracellular matrix and hormones. Melatonin, produced by the pineal gland, is a hormone that regulates sleep cycles. Recent studies have shown that melatonin improves the therapeutic effects of stem cells. The present study aimed to investigate whether melatonin enhances the biological activities of human adipose-derived MSCs. The results demonstrated that treatment with melatonin promoted cell proliferation by inducing SRY-box transcription factor 2 gene expression and preventing replicative senescence. In addition, melatonin exerted anti-adipogenic effects on MSCs. PCR analysis revealed that the expression of the CCAAT enhancer binding protein a gene, a key transcription factor in adipogenesis, was decreased following melatonin treatment, resulting in reduced adipogenic differentiation in an in vitro assay. The present study also examined the effect of melatonin on the immunomodulatory response using a co-culture system of human peripheral blood mononuclear cells and MSCs. Activated T cells were strongly inhibited following melatonin exposure compared with those in the control group. Finally, the favorable effects of melatonin on MSCs were confirmed using luzindole, a selective melatonin receptor antagonist. The proliferation-promoting, anti-inflammatory effects of melatonin suggested that melatonin-treated MSCs may be used for effective cell therapy.
Collapse
Affiliation(s)
- June Seok Heo
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Sangshin Pyo
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Ja-Yun Lim
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Dae Wui Yoon
- Department of Biomedical Laboratory Science, College of Health Science, Jungwon University, Geosan, Chungbuk 28024, Republic of Korea
| | - Bo Yong Kim
- Department of Health and Environmental Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Jin-Hee Kim
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju, North Chungcheong 28497, Republic of Korea
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Seung Gwan Lee
- Department of Health and Environmental Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Jinkwan Kim
- Department of Biomedical Laboratory Science, College of Health Science, Jungwon University, Geosan, Chungbuk 28024, Republic of Korea
| |
Collapse
|
31
|
González-González A, García Nieto E, González A, Sánchez-Fernández C, Alonso-González C, Menéndez-Menéndez J, Gómez-Arozamena J, Cos S, Martínez-Campa C. Melatonin Modulation of Radiation and Chemotherapeutics-induced Changes on Differentiation of Breast Fibroblasts. Int J Mol Sci 2019; 20:ijms20163935. [PMID: 31412584 PMCID: PMC6719206 DOI: 10.3390/ijms20163935] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/05/2019] [Accepted: 08/10/2019] [Indexed: 02/07/2023] Open
Abstract
Melatonin exerts oncostatic actions and sensitizes tumor cells to chemotherapeutics or radiation. In our study, we investigated the effects of docetaxel, vinorelbine, and radiation on human breast fibroblasts and its modulation by melatonin. Docetaxel or vinorelbine inhibits proliferation and stimulates the differentiation of breast preadipocytes, by increasing C/EBPα and PPARγ expression and by downregulating tumor necrosis factor α (TNFα), interleukin 6 (IL-6), and IL-11 expression. Radiation inhibits both proliferation and differentiation through the downregulation of C/EBPα and PPARγ and by stimulating TNFα expression. In addition, docetaxel and radiation decrease aromatase activity and expression by decreasing aromatase promoter II and cyclooxygenases 1 and 2 (COX-1 and COX-2) expression. Melatonin potentiates the stimulatory effect of docetaxel and vinorelbine on differentiation and their inhibitory effects on aromatase activity and expression, by increasing the stimulatory effect on C/EBPα and PPARγ expression and the downregulation of antiadipogenic cytokines and COX expression. Melatonin also counteracts the inhibitory effect of radiation on differentiation of preadipocytes, by increasing C/EBPα and PPARγ expression and by decreasing TNFα expression. Melatonin also potentiates the inhibitory effect exerted by radiation on aromatase activity and expression by increasing the downregulation of promoter II, and COX-1 and COX-2 expression. Our findings suggest that melatonin modulates regulatory effects induced by chemotherapeutic drugs or radiation on preadipocytes, which makes it a promising adjuvant for chemotherapy and radiotherapy sensibilization.
Collapse
Affiliation(s)
- Alicia González-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Enrique García Nieto
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain.
| | - Cristina Sánchez-Fernández
- Department of Anatomy and Cellular Biology, School of Medicine, University of Cantabria, 39011 Santander, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain.
| | - Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | - José Gómez-Arozamena
- Department of Medical Physics, School of Medicine, University of Cantabria, 39011 Santander, Spain
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| |
Collapse
|
32
|
Liu K, Yu W, Wei W, Zhang X, Tian Y, Sherif M, Liu X, Dong C, Wu W, Zhang L, Chen J. Melatonin reduces intramuscular fat deposition by promoting lipolysis and increasing mitochondrial function. J Lipid Res 2019; 60:767-782. [PMID: 30552289 PMCID: PMC6446696 DOI: 10.1194/jlr.m087619] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/14/2018] [Indexed: 01/06/2023] Open
Abstract
In obesity and diabetes, intramuscular fat (IMF) content correlates markedly with insulin sensitivity, which makes IMF manipulation an area of therapeutic interest. Melatonin, an important circadian rhythm-regulating hormone, reportedly regulates fat deposition, but its effects on different types of adipose vary. Little is known about the role of melatonin in IMF deposition. Here, using intramuscular preadipocytes in pigs, we investigated to determine whether melatonin affects or regulates IMF deposition. We found that melatonin greatly inhibited porcine intramuscular preadipocyte proliferation. Although melatonin administration significantly upregulated the expression of adipogenic genes, smaller lipid droplets were formed in intramuscular adipocytes. Additional investigation demonstrated that melatonin promoted lipolysis of IMF by activating protein kinase A and the signaling of ERK1/2. Moreover, melatonin increased thermogenesis in intramuscular adipocytes by enhancing mitochondrial biogenesis and mitochondrial respiration. A mouse model, in which untreated controls were compared with mice that received 3 weeks of melatonin treatment, verified the effect of melatonin on IMF deposition. In conclusion, melatonin reduces IMF deposition by upregulating lipolysis and mitochondrial bioactivities. These data establish a link between melatonin signaling and lipid metabolism in mammalian models and suggest the potential for melatonin administration to treat or prevent obesity and related diseases.
Collapse
Affiliation(s)
- Kaiqing Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wensai Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xinbao Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ye Tian
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Melak Sherif
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chao Dong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangjun Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
33
|
de Farias TDSM, Cruz MM, de Sa RCDC, Severi I, Perugini J, Senzacqua M, Cerutti SM, Giordano A, Cinti S, Alonso-Vale MIC. Melatonin Supplementation Decreases Hypertrophic Obesity and Inflammation Induced by High-Fat Diet in Mice. Front Endocrinol (Lausanne) 2019; 10:750. [PMID: 31749764 PMCID: PMC6848267 DOI: 10.3389/fendo.2019.00750] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity results from critical periods of positive energy balance characterized by caloric intake greater than energy expenditure. This disbalance promotes adipose tissue dysfunction which is related to other comorbidities. Melatonin is a low-cost therapeutic agent and studies indicate that its use may improve obesity-related disorders. To evaluate if the melatonin is efficient in delaying or even blocking the damages caused by excessive ingestion of a high-fat diet (HFD) in mice, as well as improving the inflammatory profile triggered by obesity herein, male C57BL/6 mice of 8 weeks were induced to obesity by a HFD and treated for 10 weeks with melatonin. The results demonstrate that melatonin supplementation attenuated serum triglyceride levels and total and LDL cholesterol and prevented body mass gain through a decreased lipogenesis rate and increased lipolytic capacity in white adipocytes, with a concomitant increment in oxygen consumption and Pgc1a and Prdm16 expression. Altogether, these effects prevented adipocyte hypertrophy caused by HFD and reflected in decreased adiposity. Finally, melatonin supplementation reduced the crown-like-structure (CLS) formation, characteristic of the inflammatory process by macrophage infiltration into white adipose tissue of obese subjects, as well as decreased the gene expression of inflammation-related factors, such as leptin and MCP1. Thus, the melatonin can be considered a potential therapeutic agent to attenuate the metabolic and inflammatory disorders triggered by obesity.
Collapse
Affiliation(s)
- Talita da Silva Mendes de Farias
- Post-graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo-UNIFESP, Diadema, Brazil
| | - Maysa Mariana Cruz
- Post-graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo-UNIFESP, Diadema, Brazil
| | - Roberta Cavalcante da Cunha de Sa
- Post-graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo-UNIFESP, Diadema, Brazil
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, University of Ancona (Politecnica Delle Marche), Ancona, Italy
- Center of Obesity, University of Ancona (Politecnica Delle Marche), Ancona, Italy
| | - Jessica Perugini
- Department of Experimental and Clinical Medicine, University of Ancona (Politecnica Delle Marche), Ancona, Italy
- Center of Obesity, University of Ancona (Politecnica Delle Marche), Ancona, Italy
| | - Martina Senzacqua
- Department of Experimental and Clinical Medicine, University of Ancona (Politecnica Delle Marche), Ancona, Italy
- Center of Obesity, University of Ancona (Politecnica Delle Marche), Ancona, Italy
| | - Suzete Maria Cerutti
- Post-graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo-UNIFESP, Diadema, Brazil
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo-UNIFESP, Diadema, Brazil
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, University of Ancona (Politecnica Delle Marche), Ancona, Italy
- Center of Obesity, University of Ancona (Politecnica Delle Marche), Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, University of Ancona (Politecnica Delle Marche), Ancona, Italy
- Center of Obesity, University of Ancona (Politecnica Delle Marche), Ancona, Italy
| | - Maria Isabel Cardoso Alonso-Vale
- Post-graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo-UNIFESP, Diadema, Brazil
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo-UNIFESP, Diadema, Brazil
- *Correspondence: Maria Isabel Cardoso Alonso-Vale
| |
Collapse
|
34
|
Tindall AM, Petersen KS, Lamendella R, Shearer GC, Murray-Kolb LE, Proctor DN, Kris-Etherton PM. Tree Nut Consumption and Adipose Tissue Mass: Mechanisms of Action. Curr Dev Nutr 2018; 2:nzy069. [PMID: 30488045 PMCID: PMC6252345 DOI: 10.1093/cdn/nzy069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/18/2018] [Accepted: 08/02/2018] [Indexed: 01/03/2023] Open
Abstract
There is concern that tree nuts may cause weight gain due to their energy density, yet evidence shows that tree nuts do not adversely affect weight status. Epidemiologic and experimental studies have shown a reduced risk of chronic diseases with tree nut consumption without an increased risk of weight gain. In fact, tree nuts may protect against weight gain and benefit weight-loss interventions. However, the relation between tree nut consumption and adiposity is not well understood at the mechanistic level. This review summarizes the proposed underlying mechanisms that might account for this relation. Evidence suggests that tree nuts may affect adiposity through appetite control, displacement of unfavorable nutrients, increased diet-induced thermogenesis, availability of metabolizable energy, antiobesity action of bioactive compounds, and improved functionality of the gut microbiome. The gut microbiome is a common factor among these mechanisms and may mediate, in part, the relation between tree nut consumption and reduced adiposity. Further research is needed to understand the impact of tree nuts on the gut microbiome and how the gut microbial environment affects the nutrient absorption and metabolism of tree nuts. The evidence to date suggests that tree nut consumption favorably affects body composition through different mechanisms that involve the gut microbiome. A better understanding of these mechanisms will contribute to the evolving science base that addresses the causes and treatments for overweight and obesity.
Collapse
Affiliation(s)
- Alyssa M Tindall
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - Kristina S Petersen
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | | | - Gregory C Shearer
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - Laura E Murray-Kolb
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - David N Proctor
- Departments of Kinesiology, The Pennsylvania State University, University Park, PA
| | - Penny M Kris-Etherton
- Departments of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| |
Collapse
|
35
|
Liu Y, Li LN, Guo S, Zhao XY, Liu YZ, Liang C, Tu S, Wang D, Li L, Dong JZ, Gao L, Yang HB. Melatonin improves cardiac function in a mouse model of heart failure with preserved ejection fraction. Redox Biol 2018; 18:211-221. [PMID: 30031269 PMCID: PMC6076208 DOI: 10.1016/j.redox.2018.07.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/06/2018] [Accepted: 07/10/2018] [Indexed: 01/06/2023] Open
Abstract
Melatonin has been shown to inhibit myocardial infarction-induced apoptosis, its function in heart failure with preserved ejection fraction (HFpEF) has not been investigated. This study aimed to investigate whether melatonin attenuates obesity-related HFpEF. Male mice were fed a high-fat diet (HFD) from weaning to 6 months of age to induce HFpEF. The mice were orally administered melatonin (50 mg/kg) by 3 weeks. Diastolic function was significantly improved by melatonin supplementation in mice fed an HFD. Melatonin attenuated obesity-induced myocardial oxidative stress and apoptosis and promoted the secretion of C1q/tumour necrosis factor-related protein 3 (CTRP3) by adipose tissue. And depletion of circulating CTRP3 largely abolished melatonin-mediated cardio-protection. Melatonin-mediated secretion of adipocyte-derived CTRP3 activated NF-E2-related factor 2 (Nrf2), which were largely abrogated by knocking down CTRP3 in adipocytes or Nrf2 in cardiomyocytes. Nrf2 activation was mediated by miR-200a, and a miR-200a antagomir offset the effects of melatonin-conditioned medium on Nrf2 expression. Our results indicate that melatonin can be used to treat and prevent obesity-related HFpEF. Melatonin attenuates obesity-related heart failure with preserved ejection fraction. Melatonin attenuates obesity-induced myocardial injury via adipose tissue-derived CTRP3. Melatonin-mediated secretion of adipocyte-derived CTRP3 activates Nrf2 by miR-200a.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li-Na Li
- Department of Anaesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sen Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Yan Zhao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu-Zhou Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cui Liang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sheng Tu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dan Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ling Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian-Zeng Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Hai-Bo Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
36
|
Unno Y, Yamamoto H, Takatsuki S, Sato Y, Kuranaga T, Yazawa K, Ono Y, Wakimoto T. Palmitoyl lactic acid induces adipogenesis and a brown fat-like phenotype in 3T3-L1 preadipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:772-782. [DOI: 10.1016/j.bbalip.2018.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/26/2018] [Accepted: 04/08/2018] [Indexed: 12/01/2022]
|
37
|
Fernández Vázquez G, Reiter RJ, Agil A. Melatonin increases brown adipose tissue mass and function in Zücker diabetic fatty rats: implications for obesity control. J Pineal Res 2018; 64:e12472. [PMID: 29405372 DOI: 10.1111/jpi.12472] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 01/19/2018] [Indexed: 12/30/2022]
Abstract
Melatonin limits obesity in rodents without affecting food intake and activity, suggesting a thermogenic effect. Previously we demonstrated that melatonin browns subcutaneous fat in Zücker diabetic fatty (ZDF) rats. Other works pointed to melatonin as a signal that increases brown adipose tissue (BAT) mass and function in rodents. However, direct proof of thermogenic properties (uncoupled mitochondria) of the newly recruited BAT in response to melatonin is still lacking. Therefore, in this work, we investigated if melatonin recruits thermogenic BAT in ZDF rats. Zücker lean (ZL) and ZDF animals were subdivided into two groups, control (C) and treated with oral melatonin (M) for 6 weeks. Mitochondrial mass, activity of citrate synthase (CS), and respiratory chain complexes I and IV were lower in C-ZDF than in C-ZL animals (P < .001). Melatonin treatment increased BAT weight in ZDF rats (P < .001). Also, it rose mitochondrial mass (P < .01) and activities of CS and complexes I and IV (P < .001) in both, ZDF and ZL rats. Uncoupling protein 1 (UCP1) mRNA and protein were 50% lower in BAT from obese rats. Also, guanosine diphosphate (GDP) binding was lower in ZDF than in lean rats (P < .01). Melatonin treatment of obese rats restored the expression of UCP1 and GDP binding to levels of lean rats and sensitized the thermogenic response to cold exposure. These data demonstrated that melatonin recruits thermogenic BAT in ZDF rats. This may contribute to melatonin's control of body weight and its metabolic benefits.
Collapse
Affiliation(s)
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science at San Antonio, San Antonio, TX, USA
| | - Ahmad Agil
- Deparment of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
38
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Demyelination in Multiple Sclerosis: Reprogramming Energy Metabolism and Potential PPARγ Agonist Treatment Approaches. Int J Mol Sci 2018; 19:ijms19041212. [PMID: 29659554 PMCID: PMC5979570 DOI: 10.3390/ijms19041212] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 12/20/2022] Open
Abstract
Demyelination in multiple sclerosis (MS) cells is the site of several energy metabolic abnormalities driven by dysregulation between the opposed interplay of peroxisome proliferator-activated receptor γ (PPARγ) and WNT/β-catenin pathways. We focus our review on the opposing interactions observed in demyelinating processes in MS between the canonical WNT/β-catenin pathway and PPARγ and their reprogramming energy metabolism implications. Demyelination in MS is associated with chronic inflammation, which is itself associated with the release of cytokines by CD4+ Th17 cells, and downregulation of PPARγ expression leading to the upregulation of the WNT/β-catenin pathway. Upregulation of WNT/β-catenin signaling induces activation of glycolytic enzymes that modify their energy metabolic behavior. Then, in MS cells, a large portion of cytosolic pyruvate is converted into lactate. This phenomenon is called the Warburg effect, despite the availability of oxygen. The Warburg effect is the shift of an energy transfer production from mitochondrial oxidative phosphorylation to aerobic glycolysis. Lactate production is correlated with increased WNT/β-catenin signaling and demyelinating processes by inducing dysfunction of CD4+ T cells leading to axonal and neuronal damage. In MS, downregulation of PPARγ decreases insulin sensitivity and increases neuroinflammation. PPARγ agonists inhibit Th17 differentiation in CD4+ T cells and then diminish release of cytokines. In MS, abnormalities in the regulation of circadian rhythms stimulate the WNT pathway to initiate the demyelination process. Moreover, PPARγ contributes to the regulation of some key circadian genes. Thus, PPARγ agonists interfere with reprogramming energy metabolism by directly inhibiting the WNT/β-catenin pathway and circadian rhythms and could appear as promising treatments in MS due to these interactions.
Collapse
Affiliation(s)
- Alexandre Vallée
- Délégation à la Recherche Clinique et à l'Innovation (DRCI), Hôpital Foch, 92150 Suresnes, France.
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), 77100 Meaux, France.
| | - Rémy Guillevin
- Data Analysis and Computations Through Imaging Modeling-Mathématiques (DACTIM), Unité mixte de recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 7348 (Laboratoire de Mathématiques et Application), University of Poitiers, Centre Hospitalier Universitaire (CHU) de Poitiers, 86000 Poitiers, France.
| | - Jean-Noël Vallée
- Centre Hospitalier Universitaire (CHU) Amiens Picardie, University of Picardie Jules Verne (UPJV), 80000 Amiens, France.
- LMA (Laboratoire de Mathématiques et Applications), Unité mixte de recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 7348, Université de Poitiers, 86000 Poitiers, France.
| |
Collapse
|
39
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Thermodynamics in Neurodegenerative Diseases: Interplay Between Canonical WNT/Beta-Catenin Pathway-PPAR Gamma, Energy Metabolism and Circadian Rhythms. Neuromolecular Med 2018; 20:174-204. [PMID: 29572723 DOI: 10.1007/s12017-018-8486-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 03/20/2018] [Indexed: 02/06/2023]
Abstract
Entropy production rate is increased by several metabolic and thermodynamics abnormalities in neurodegenerative diseases (NDs). Irreversible processes are quantified by changes in the entropy production rate. This review is focused on the opposing interactions observed in NDs between the canonical WNT/beta-catenin pathway and PPAR gamma and their metabolic and thermodynamic implications. In amyotrophic lateral sclerosis and Huntington's disease, WNT/beta-catenin pathway is upregulated, whereas PPAR gamma is downregulated. In Alzheimer's disease and Parkinson's disease, WNT/beta-catenin pathway is downregulated while PPAR gamma is upregulated. The dysregulation of the canonical WNT/beta-catenin pathway is responsible for the modification of thermodynamics behaviors of metabolic enzymes. Upregulation of WNT/beta-catenin pathway leads to aerobic glycolysis, named Warburg effect, through activated enzymes, such as glucose transporter (Glut), pyruvate kinase M2 (PKM2), pyruvate dehydrogenase kinase 1(PDK1), monocarboxylate lactate transporter 1 (MCT-1), lactic dehydrogenase kinase-A (LDH-A) and inactivation of pyruvate dehydrogenase complex (PDH). Downregulation of WNT/beta-catenin pathway leads to oxidative stress and cell death through inactivation of Glut, PKM2, PDK1, MCT-1, LDH-A but activation of PDH. In addition, in NDs, PPAR gamma is dysregulated, whereas it contributes to the regulation of several key circadian genes. NDs show many dysregulation in the mediation of circadian clock genes and so of circadian rhythms. Thermodynamics rhythms operate far-from-equilibrium and partly regulate interactions between WNT/beta-catenin pathway and PPAR gamma. In NDs, metabolism, thermodynamics and circadian rhythms are tightly interrelated.
Collapse
Affiliation(s)
- Alexandre Vallée
- DRCI, Hôpital Foch, Suresnes, France.
- LMA (Laboratoire de Mathématiques et Applications) CNRS 7348, University of Poitiers, 11 Boulevard Marie et Pierre Curie, Poitiers, France.
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, Université de Poitiers et CHU de Poitiers, Poitiers, France
| | - Jean-Noël Vallée
- DRCI, Hôpital Foch, Suresnes, France
- CHU Amiens Picardie, Université Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
40
|
Kato H, Masuda S, Ohira T, Ohira L, Takakura H, Ohira Y, Izawa T. Differential response of adipose tissue gene and protein expressions to 4- and 8-week administration of β-guanidinopropionic acid in mice. Physiol Rep 2018; 6:e13616. [PMID: 29512301 PMCID: PMC5840394 DOI: 10.14814/phy2.13616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 11/24/2022] Open
Abstract
β-Guanidinopropionic acid (β-GPA) feeding inhibits growth-associated gain of body mass. It remains unknown, however, whether and how β-GPA feeding affects growth-associated increase in white adipose tissue (WAT) mass. We examined the effects of 4- and 8-week β-GPA feeding on serum myostatin levels and expression of genes and proteins related to adipogenesis, lipolysis, and liposynthesis in epididymal WAT (eWAT) and brown adipose tissue (BAT) in 3-week-old, juvenile male mice. Body, eWAT, and muscle weights were significantly lower in β-GPA-fed mice than in controls after feeding. Four- but not 8-week-β-GPA feeding increased the serum myostatin level. Incubation of C2C12 myotubes with β-GPA (1 mM) significantly promoted myostatin mRNA expression. The protein expression of peroxisome proliferator-activated receptor gamma coactivator 1 α (PGC-1α) and peroxisome proliferator-activated receptor α (PPARα) was up-regulated in GPAF eWAT at week 4, but down-regulated at week 8. There was no significant difference in the protein expression of adipocyte triglyceride lipase (ATGL), hormone-sensitive lipase (HSL), fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC) between groups in eWAT. In BAT, no significant difference was found in the protein expression of PGC-1α, PPARα, ATGL, and HSL between β-GPA-fed and control mice, whereas that of FAS and ACC was significantly lower in β-GPA-fed mice at week 8. Uncoupling protein 1 was expressed higher in β-GPA-fed mice both at weeks 4 and 8 than that in controls. Thus, the mechanism by which β-GPA feeding in early juvenile mice inhibits growth-associated increase in eWAT mass may differ between early and later periods of growth.
Collapse
Affiliation(s)
- Hisashi Kato
- Faculty and Graduate School of Health and Sports ScienceDoshisha UniversityKyotanabeJapan
| | - Shinya Masuda
- Faculty and Graduate School of Health and Sports ScienceDoshisha UniversityKyotanabeJapan
| | | | - Luna Ohira
- Faculty of Health and Well‐beingKansai UniversitySakaiJapan
| | - Hisashi Takakura
- Faculty and Graduate School of Health and Sports ScienceDoshisha UniversityKyotanabeJapan
- Research Center for Adipocyte and Muscle ScienceDoshisha UniversityKyotanabeJapan
| | - Yoshinobu Ohira
- Faculty and Graduate School of Health and Sports ScienceDoshisha UniversityKyotanabeJapan
- Research Center for Adipocyte and Muscle ScienceDoshisha UniversityKyotanabeJapan
- Research Center for Space Medical ScienceDoshisha UniversityKyotanabeJapan
- Graduate School of MedicineOsaka UniversityToyonakaJapan
| | - Tetsuya Izawa
- Faculty and Graduate School of Health and Sports ScienceDoshisha UniversityKyotanabeJapan
- Research Center for Adipocyte and Muscle ScienceDoshisha UniversityKyotanabeJapan
- Research Center for Space Medical ScienceDoshisha UniversityKyotanabeJapan
| |
Collapse
|
41
|
Valenzuela-Melgarejo FJ, Caro-Díaz C, Cabello-Guzmán G. Potential Crosstalk between Fructose and Melatonin: A New Role of Melatonin-Inhibiting the Metabolic Effects of Fructose. Int J Endocrinol 2018; 2018:7515767. [PMID: 30154843 PMCID: PMC6092995 DOI: 10.1155/2018/7515767] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/22/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Increased consumption of energy-dense foods such as fructose-rich syrups represents one of the significant, growing concerns related to the alarming trend of overweight, obesity, and metabolic disorders worldwide. Metabolic pathways affected by fructose involve genes related to lipogenesis/lipolysis, beta-oxidation, mitochondrial biogenesis, gluconeogenesis, oxidative phosphorylation pathways, or altering of circadian production of insulin and leptin. Moreover, fructose can be a risk factor during pregnancy elevating the risk of preterm delivery, hypertension, and metabolic impairment of the mother and fetus. Melatonin is a chronobiotic and homeostatic hormone that can modulate the harmful effects of fructose via clock gene expression and metabolic pathways, modulating the expression of PPARγ, SREBF-1 (SREBP-1), hormone-sensitive lipase, C/EBP-α genes, NRF-1, PGC1α, and uncoupling protein-1. Moreover, this hormone has the capacity in the rat of reverting the harmful effects of fructose, increasing the body weight and weight ratio of the liver, and increasing the body weight and restoring the glycemia from mothers exposed to fructose. The aim of this review is to show the potential crosstalk between fructose and melatonin and their potential role during pregnancy.
Collapse
Affiliation(s)
| | - Claudia Caro-Díaz
- Laboratory of Molecular Cell Biology, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Chillán, Chile
| | - Gerardo Cabello-Guzmán
- Laboratory of Molecular Cell Biology, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Chillán, Chile
| |
Collapse
|
42
|
Liu Z, Gan L, Zhang T, Ren Q, Sun C. Melatonin alleviates adipose inflammation through elevating α-ketoglutarate and diverting adipose-derived exosomes to macrophages in mice. J Pineal Res 2018; 64. [PMID: 29149454 DOI: 10.1111/jpi.12455] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/06/2017] [Indexed: 12/21/2022]
Abstract
Obesity is associated with macrophage infiltration and metabolic inflammation, both of which promote metabolic disease progression. Melatonin is reported to possess anti-inflammatory properties by inhibiting inflammatory response of adipocytes and macrophages activation. However, the effects of melatonin on the communication between adipocytes and macrophages during adipose inflammation remain elusive. Here, we demonstrated melatonin alleviated inflammation and elevated α-ketoglutarate (αKG) level in adipose tissue of obese mice. Mitochondrial isocitrate dehydrogenase 2 (Idh2) mRNA level was also elevated by melatonin in adipocytes leading to increase αKG level. Further analysis revealed αKG was the target for melatonin inhibition of adipose inflammation. Moreover, sirtuin 1 (Sirt1) physically interacted with IDH2 and formed a complex to increase the circadian amplitude of Idh2 and αKG content in melatonin-inhibited adipose inflammation. Notably, melatonin promoted exosomes secretion from adipocyte and increased adipose-derived exosomal αKG level. Our results also confirmed that melatonin alleviated adipocyte inflammation and increased ratio of M2 to M1 macrophages by transporting of exosomal αKG to macrophages and promoting TET-mediated DNA demethylation. Furthermore, exosomal αKG attenuated signal transducers and activators of transduction-3 (STAT3)/NF-κB signal by its receptor oxoglutarate receptor 1 (OXGR1) in adipocytes. Melatonin also attenuated adipose inflammation and deceased macrophage number in chronic jet-lag mice. In summary, our results demonstrate melatonin alleviates metabolic inflammation by increasing cellular and exosomal αKG level in adipose tissue. Our data reveal a novel function of melatonin on adipocytes and macrophages communication, suggesting a new potential therapy for melatonin to prevent and treat obesity caused systemic inflammatory disease.
Collapse
Affiliation(s)
- Zhenjiang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Lu Gan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Tiantian Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Qian Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
43
|
Melatonin promotes triacylglycerol accumulation via MT2 receptor during differentiation in bovine intramuscular preadipocytes. Sci Rep 2017; 7:15080. [PMID: 29118419 PMCID: PMC5678110 DOI: 10.1038/s41598-017-12780-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/14/2017] [Indexed: 12/22/2022] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is a derivative of tryptophan which is produced and secreted mainly by the pineal gland and regulates a variety of important central and peripheral actions. To examine the potential effects of melatonin on the proliferation and differentiation of bovine intramuscular preadipocytes (BIPs), BIPs were incubated with different concentrations of melatonin. Melatonin supplementation at 1 mM significantly increased peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer-binding protein (C/EBP) β, and C/EBPα expression and promoted the differentiation of BIPs into adipocytes with large lipid droplets and high cellular triacylglycerol (TAG) levels. Melatonin also significantly enhanced lipolysis and up-regulated the expression of lipolytic genes and proteins, including hormone sensitive lipase (HSL), adipocyte triglyceride lipase (ATGL), and perilipin 1 (PLIN1). Moreover, melatonin reduced intracellular reactive oxygen species (ROS) levels by increasing the expression levels and activities of superoxide dismutase 1 (SOD1) and glutathione peroxidase 4 (GPX4). Finally, the positive effects of melatonin on adipogenesis, lipolysis, and redox status were reversed by treatment with luzindole, anantagonist of nonspecific melatonin receptors 1 (MT1) and 2 (MT2), and 4-phenyl-2-propionamidotetraline (4P-PDOT), a selective MT2 antagonist. These results reveal that melatonin promotes TAG accumulation via MT2 receptor during differentiation in BIPs.
Collapse
|
44
|
Reprogramming energetic metabolism in Alzheimer's disease. Life Sci 2017; 193:141-152. [PMID: 29079469 DOI: 10.1016/j.lfs.2017.10.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/23/2017] [Indexed: 12/20/2022]
Abstract
Entropy rate is increased by several metabolic and thermodynamics abnormalities in neurodegenerative diseases (NDs). Changes in Gibbs energy, heat production, ionic conductance or intracellular acidity are irreversibles processes which driven modifications of the entropy rate. The present review focusses on the thermodynamic implications in the reprogramming of cellular energy metabolism enabling in Alzheimer's disease (AD) through the opposite interplay of the molecular signaling pathways WNT/β-catenin and PPARγ. In AD, WNT/β-catenin pathway is downregulated while PPARγ is upregulated. Thermodynamics behaviors of metabolic enzymes are modified by dysregulation of the canonical WNT/β-catenin pathway. Downregulation of WNT/β-catenin pathway leads to oxidative stress and cell death through inactivation of glycolytic enzymes such as Glut, PKM2, PDK1, MCT-1, LDH-A but activation of PDH. In addition, in NDs, PPARγ is dysregulated whereas it contributes to the regulation of several key circadian genes. AD is considered as a dissipative structure that exchanges energy or matter with its environment far from the thermodynamic equilibrium. Far-from-equilibrium thermodynamics are notions driven by circadian rhythms. Circadian rhythms directly participate in regulating the molecular pathways WNT/β-catenin and PPARγ involved in the reprogramming of cellular energy metabolism enabling AD processes.
Collapse
|
45
|
Crooke A, Huete-Toral F, Colligris B, Pintor J. The role and therapeutic potential of melatonin in age-related ocular diseases. J Pineal Res 2017; 63. [PMID: 28658514 DOI: 10.1111/jpi.12430] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/23/2017] [Indexed: 12/20/2022]
Abstract
The eye is continuously exposed to solar UV radiation and pollutants, making it prone to oxidative attacks. In fact, oxidative damage is a major cause of age-related ocular diseases including cataract, glaucoma, age-related macular degeneration, and diabetic retinopathy. As the nature of lens cells, trabecular meshwork cells, retinal ganglion cells, retinal pigment epithelial cells, and photoreceptors is postmitotic, autophagy plays a critical role in their cellular homeostasis. In age-related ocular diseases, this process is impaired, and thus, oxidative damage becomes irreversible. Other conditions such as low-grade chronic inflammation and angiogenesis also contribute to the development of retinal diseases (glaucoma, age-related macular degeneration and diabetic retinopathy). As melatonin is known to have remarkable qualities such as antioxidant/antinitridergic, mitochondrial protector, autophagy modulator, anti-inflammatory, and anti-angiogenic, it can represent a powerful tool to counteract all these diseases. The present review analyzes the role and therapeutic potential of melatonin in age-related ocular diseases, focusing on nitro-oxidative stress, autophagy, inflammation, and angiogenesis mechanisms.
Collapse
Affiliation(s)
- Almudena Crooke
- Department of Biochemistry and Molecular Biology IV, Group Ocupharm, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Fernando Huete-Toral
- Department of Biochemistry and Molecular Biology IV, Group Ocupharm, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Basilio Colligris
- Department of Biochemistry and Molecular Biology IV, Group Ocupharm, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Jesús Pintor
- Department of Biochemistry and Molecular Biology IV, Group Ocupharm, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
46
|
Kokune-Takahashi A, Ayaki M, Tsubota K, Negishi K. Effects of Cataract Opacity and Surgery on Sleep Quality. Rejuvenation Res 2017; 21:53-60. [PMID: 28728538 DOI: 10.1089/rej.2016.1907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This study aimed to identify the type of cataract opacity associated with subjective sleep quality for cataract patients. A total of 180 consecutive patients (average age 74.2 years) underwent cataract surgery with implantation of an ultraviolet-blocking or blue light-blocking intraocular lens. The participants' subjective sleep quality was evaluated using the Pittsburgh sleep quality index (PSQI) before and at 2 months after surgery. Patients were divided into two groups for analysis: normal sleepers (preoperative PSQI ≤5) and poor sleepers (preoperative PSQI ≥6). The preoperative and postoperative PSQI scores were 2.8 ± 1.5 and 3.2 ± 2.0, respectively, for normal sleepers (n = 99), and 8.5 ± 2.9 and 7.4 ± 3.3, respectively, for poor sleepers (n = 81). The improvement in PSQI was significant in poor sleepers (p < 0.0001, Wilcoxon's test) with measured values of 0.22 ± 0.96 hours extension in sleep duration and 0.16 ± 0.62 hours shortening in sleep latency. Patients with posterior subcapsular cataract (PSC) and nuclear opacity showed the greatest improvement in PSQI score, with regression analysis identifying PSC as having a significant effect on improvement in PSQI. Significant correlations were found between the subjective sleep and cataract opacity in cataract patients. Ophthalmic personnel should therefore be aware that cataract patients with PSC have a greater potential for disability and predictable benefits from surgery in vision and subjective sleep.
Collapse
Affiliation(s)
| | - Masahiko Ayaki
- Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine , Tokyo, Japan
| |
Collapse
|
47
|
Chaudhary S, Parvez S. Phytanic acid induced neurological alterations in rat brain synaptosomes and its attenuation by melatonin. Biomed Pharmacother 2017; 95:37-46. [PMID: 28826095 DOI: 10.1016/j.biopha.2017.07.156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/21/2017] [Accepted: 07/30/2017] [Indexed: 12/17/2022] Open
Abstract
Phytanic acid (3,7,11,15-tetramethylhexadecanoic acid) (Phyt) is a saturated branched chain fatty acid which originates after the breakdown of chlorophyll molecule, phytol. It plays an important role in a variety of metabolic disorders with peroxisomal impairments. The aim of our investigation was to evaluate the adverse effects of Phyt on synaptic functions by using synaptosomal preparation of rat brain as an in vitro model and the possible protective role of melatonin against Phyt-induced neurotoxicity. Melatonin is an antioxidant, secreted by the pineal gland. Melatonin and its metabolites have neuroprotective effects on cellular stress, by reducing reactive oxygen species (ROS) and reactive nitrogen species (RNS). In the present investigation, synaptosomes prepared from rat brain were co-treated with melatonin (10μM) and Phyt (50μM) for 2h. Co-treatment of Phyt with melatonin significantly restored the altered levels of protein carbonyl (PC) contents and lipid peroxidation (LPO). It also replenished the Phyt-induced alterations on the levels of non-enzymatic antioxidant defence reduced glutathione (GSH), enzymatic antioxidants such as catalase (CAT) and superoxide dismutase (SOD) and synaptosomal integral enzymes such as AChE, Na+, K+-ATPase and MAO. We observed that Phyt induced oxidative stress in synaptosomes as indicated by an elevation in the generation of ROS and melatonin was able to inhibit the elevated ROS generation. Moreover, the neurotoxic effects elicited by Phyt on NO level and membrane potential were totally prevented by the treatment of melatonin. The results of our investigation emphasize the potential use of melatonin as a nutraceutical and mitigatory agent against Phyt-induced oxidative stress.
Collapse
Affiliation(s)
- Shaista Chaudhary
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India.
| |
Collapse
|
48
|
Melatonin Decreases Glucose Metabolism in Prostate Cancer Cells: A 13C Stable Isotope-Resolved Metabolomic Study. Int J Mol Sci 2017; 18:ijms18081620. [PMID: 28933733 PMCID: PMC5578012 DOI: 10.3390/ijms18081620] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 12/18/2022] Open
Abstract
The pineal neuroindole melatonin exerts an exceptional variety of systemic functions. Some of them are exerted through its specific membrane receptors type 1 and type 2 (MT1 and MT2) while others are mediated by receptor-independent mechanisms. A potential transport of melatonin through facilitative glucose transporters (GLUT/SLC2A) was proposed in prostate cancer cells. The prostate cells have a particular metabolism that changes during tumor progression. During the first steps of carcinogenesis, oxidative phosphorylation is reactivated while the switch to the “Warburg effect” only occurs in advanced tumors and in the metastatic stage. Here, we investigated whether melatonin might change prostate cancer cell metabolism. To do so, 13C stable isotope-resolved metabolomics in androgen sensitive LNCaP and insensitive PC-3 prostate cancer cells were employed. In addition to metabolite 13C-labeling, ATP/AMP levels, and lactate dehydrogenase or pentose phosphate pathway activity were measured. Melatonin reduces lactate labeling in androgen-sensitive cells and it also lowers 13C-labeling of tricarboxylic acid cycle metabolites and ATP production. In addition, melatonin reduces lactate 13C-labeling in androgen insensitive prostate cancer cells. Results demonstrated that melatonin limits glycolysis as well as the tricarboxylic acid cycle and pentose phosphate pathway in prostate cancer cells, suggesting that the reduction of glucose uptake is a major target of the indole in this tumor type.
Collapse
|
49
|
Ryu V, Zarebidaki E, Albers HE, Xue B, Bartness TJ. Short photoperiod reverses obesity in Siberian hamsters via sympathetically induced lipolysis and Browning in adipose tissue. Physiol Behav 2017; 190:11-20. [PMID: 28694154 DOI: 10.1016/j.physbeh.2017.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/26/2017] [Accepted: 07/06/2017] [Indexed: 10/19/2022]
Abstract
Changes in photoperiod length are transduced into neuroendocrine signals by melatonin (MEL) secreted by the pineal gland triggering seasonally adaptive responses in many animal species. Siberian hamsters, transferred from a long-day 'summer-like' photoperiod (LD) to a short-day 'winter-like' photoperiod (SD), exhibit a naturally-occurring reversal in obesity. Photoperiod-induced changes in adiposity are mediated by the duration of MEL secretion and can be mimicked by exogenously administered MEL into animals housed in LD. Evidence suggests that MEL increases the sympathetic nervous system (SNS) drive to white adipose tissue (WAT). Here, we investigated whether MEL-driven seasonally adaptive losses in body fat are associated with WAT lipolysis and browning. Hamsters were subcutaneously administered vehicle (LD+VEH) or 0.4mg/kg MEL (LD+MEL) daily for 10weeks while animals housed in SD served as a positive control. MEL and SD exposure significantly decreased the retroperitoneal (RWAT), inguinal (IWAT), epididymal (EWAT) WAT, food intake and caused testicular regression compared with the LD+VEH group. MEL/SD induced lipolysis in the IWAT and EWAT, browning of the RWAT, IWAT, and EWAT, and increased UCP1 expression in the IBAT. Additionally, MEL/SD significantly increased the number of shared MEL receptor 1a and dopamine beta-hydroxylase-immunoreactive neurons in discrete brain sites, notably the paraventricular hypothalamic nucleus, dorsomedial hypothalamic nucleus, arcuate nucleus, locus coeruleus and dorsal motor nucleus of vagus. Collectively, these findings support our hypothesis that SD-exposed Siberian hamsters undergo adaptive decreases in body adiposity due to SNS-stimulated lipid mobilization and generalized WAT browning.
Collapse
Affiliation(s)
- Vitaly Ryu
- Department of Biology, Center for Obesity Reversal, Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA.
| | - Eleen Zarebidaki
- Department of Biology, Center for Obesity Reversal, Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA
| | - H Elliott Albers
- Department of Biology, Center for Obesity Reversal, Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA
| | - Bingzhong Xue
- Department of Biology, Center for Obesity Reversal, Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA
| | - Timothy J Bartness
- Department of Biology, Center for Obesity Reversal, Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA
| |
Collapse
|
50
|
Liu Z, Gan L, Luo D, Sun C. Melatonin promotes circadian rhythm-induced proliferation through Clock/histone deacetylase 3/c-Myc interaction in mouse adipose tissue. J Pineal Res 2017; 62. [PMID: 27987529 DOI: 10.1111/jpi.12383] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 12/12/2016] [Indexed: 12/17/2022]
Abstract
Melatonin is synthesized in the pineal gland and controls circadian rhythm of peripheral adipose tissue, resulting in changes in body weight. Although core regulatory components of clock rhythmicity have been defined, insight into the mechanisms of circadian rhythm-mediated proliferation in adipose tissue is still limited. Here, we showed that melatonin (20 mg/kg/d) promoted circadian and proliferation processes in white adipose tissue. The circadian amplitudes of brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1 (Bmal1, P<.05) and circadian locomotor output cycles kaput (Clock, P<.05), period 2 (Per2, P<.05), cyclin E (P<.05), and c-Myc (P<.05) were directly increased by melatonin in adipose tissue. Melatonin also promoted cell cycle and increased cell numbers (P<.05), which was correlated with the Clock expression (P<.05). Further analysis demonstrated that Clock bound to the E-box elements in the promoter region of c-Myc and then directly stimulated c-Myc transcription. Moreover, Clock physically interacted with histone deacetylase 3 (HDAC3) and formed a complex with c-Myc to promote adipocyte proliferation. Melatonin also attenuated circadian disruption and promoted adipocyte proliferation in chronic jet-lagged mice and obese mice. Thus, our study found that melatonin promoted adipocyte proliferation by forming a Clock/HDAC3/c-Myc complex and subsequently driving the circadian amplitudes of proliferation genes. Our data reveal a novel mechanism that links circadian rhythm to cell proliferation in adipose tissue. These findings also identify a new potential means for melatonin to prevent and treat sleep deprivation-caused obesity.
Collapse
Affiliation(s)
- Zhenjiang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Lu Gan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Dan Luo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|