1
|
Chiang CH, Zhang TR, Hsu PS, Lin SP, Chen CY. Weight regain, but not weight loss exacerbates hepatic fibrosis during multiple weight cycling events in male mice. Eur J Nutr 2024; 63:965-976. [PMID: 38265751 DOI: 10.1007/s00394-024-03326-w] [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: 09/10/2023] [Accepted: 01/12/2024] [Indexed: 01/25/2024]
Abstract
PURPOSE Weight cycling is a phenomenon characterized by fluctuating body weight that is commonly observed in individuals employing intentional weight loss methods. Despite its prevalence, the impact of weight cycling on health remains equivocal. The current investigation aimed to examine the effects of weight cycling on liver health. METHODS The weight cycling model was established by switching the feeding method of mice between ad libitum (AL) and restricted intake (DR or 60% of AL) of the breeding diet to cause weight gain and weight loss, respectively. The weight cycling model comprised two and a half cycles, with one group terminating the experience during the weight-gain period (S-AL) and the other during the weight-loss period (S-DR). Liver tissue was collected to investigate morphology alterations, apoptosis, lipid metabolism, and mitochondrial homeostasis. RESULTS The results demonstrated that the termination point of weight cycling affected body weight and hepatic steatosis. All parameters examined in the S-DR mice exhibited a comparable trend to those observed in the DR mice. Notably, S-AL mice showed a significant increase in lipid metabolism-related proteins in the liver compared to AL-fed mice, along with reduced lipid droplets. Moreover, hepatic apoptosis and fibrosis were exacerbated in the S-AL mice compared to AL mice, whereas mitochondrial fusion, biogenesis, and mitophagy were decreased in the S-AL mice. CONCLUSION Weight cycling ending in weight gain exacerbated hepatic fibrosis, potentially by inducing apoptosis or disrupting mitochondrial homeostasis. Conversely, weight cycling ending in weight loss demonstrated beneficial effects on hepatic health.
Collapse
Affiliation(s)
- Chun-Hsien Chiang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Ting-Rui Zhang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Pu-Sheng Hsu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Shau-Ping Lin
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Ching-Yi Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
2
|
May MA, Tomanek L. Uncovering the roles of sirtuin activity and food availability during the onset of the heat shock response in the California mussel (Mytilus californianus): Implications for antioxidative stress responses. Comp Biochem Physiol B Biochem Mol Biol 2024; 269:110902. [PMID: 37690509 DOI: 10.1016/j.cbpb.2023.110902] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/13/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
Sirtuins are a class of NAD+-dependent deacylases, with known regulatory roles in energy metabolism and cellular stress responses in vertebrates. Previous work using marine mussels have suggested a similar role in invertebrates, providing a potential mechanism linking food availability and thermal sensitivity in Mytilids. Sirtuin inhibitors affect mussels' recovery from environmental stressors, including acute heat shock and well-fed mussels exposed to sirtuin inhibitors and/or acute heat shock respond differently than poorly fed mussels, at the protein and whole-organism levels. While this implies a relationship between sirtuins, food availability, and temperature, the direct effects of sirtuin inhibitors (nicotinamide and suramin) on sirtuin activity or their putative effectors have not been explicitly tested. In this study, adult Mytilus californianus were acclimated to a low or high food availability and exposed to one of the following treatments: control, acute heat shock, sirtuin inhibitors, or acute heat shock and sirtuin inhibitors. Mussels increased sirtuin activity during early recovery (5 h) from sirtuin inhibition and acute heat shock, but only if acclimated to a high food availability. Redox balance was also impacted in mussels acclimated to high food availability and exposed to sirtuin inhibitors, signifying interactions between ration, acute heat shock, and sirtuin inhibitors. Additionally, we found a correlation between sirtuin and superoxide dismutase activities, suggesting a potential regulatory role of oxidative stress by sirtuins. Following prolonged recovery (17 h), we found increased sirtuin activity in mussels acclimated to low food availability, indicating that endogenous sirtuin activity may be related to food availability in mussels.
Collapse
Affiliation(s)
- Melissa A May
- Florida Gulf Coast University, Fort Myers, FL 33965, USA; California Polytechnic State University, San Luis Obispo, CA 93407, USA.
| | - Lars Tomanek
- California Polytechnic State University, San Luis Obispo, CA 93407, USA
| |
Collapse
|
3
|
Geiger M, Gorica E, Mohammed SA, Mongelli A, Mengozi A, Delfine V, Ruschitzka F, Costantino S, Paneni F. Epigenetic Network in Immunometabolic Disease. Adv Biol (Weinh) 2024; 8:e2300211. [PMID: 37794610 DOI: 10.1002/adbi.202300211] [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: 06/08/2023] [Revised: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Although a large amount of data consistently shows that genes affect immunometabolic characteristics and outcomes, epigenetic mechanisms are also heavily implicated. Epigenetic changes, including DNA methylation, histone modification, and noncoding RNA, determine gene activity by altering the accessibility of chromatin to transcription factors. Various factors influence these alterations, including genetics, lifestyle, and environmental cues. Moreover, acquired epigenetic signals can be transmitted across generations, thus contributing to early disease traits in the offspring. A closer investigation is critical in this aspect as it can help to understand the underlying molecular mechanisms further and gain insights into potential therapeutic targets for preventing and treating diseases arising from immuno-metabolic dysregulation. In this review, the role of chromatin alterations in the transcriptional modulation of genes involved in insulin resistance, systemic inflammation, macrophage polarization, endothelial dysfunction, metabolic cardiomyopathy, and nonalcoholic fatty liver disease (NAFLD), is discussed. An overview of emerging chromatin-modifying drugs and the importance of the individual epigenetic profile for personalized therapeutic approaches in patients with immuno-metabolic disorders is also presented.
Collapse
Affiliation(s)
- Martin Geiger
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessandro Mengozi
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- Department of Research and Education, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| |
Collapse
|
4
|
Fedorczak A, Lewiński A, Stawerska R. Involvement of Sirtuin 1 in the Growth Hormone/Insulin-like Growth Factor 1 Signal Transduction and Its Impact on Growth Processes in Children. Int J Mol Sci 2023; 24:15406. [PMID: 37895086 PMCID: PMC10607608 DOI: 10.3390/ijms242015406] [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: 09/07/2023] [Revised: 10/01/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
The regulation of growth processes in children depends on the synthesis of growth hormone (GH) and insulin-like growth factor 1 (IGF-1). Insulin-like growth factor 1, which is mainly secreted in the liver in response to GH, is the main peripheral mediator of GH action. Newly discovered factors regulating GH secretion and its effects are being studied recently. One of them is sirtuin 1 (SIRT1). This NAD+-dependent deacetylase, by modulating the JAK2/STAT pathway, is involved in the transduction of the GH signal in hepatocytes, leading to the synthesis of IGF-1. In addition, it participates in the regulation of the synthesis of GHRH in the hypothalamus and GH in the somatotropic cells. SIRT1 is suggested to be involved in growth plate chondrogenesis and longitudinal bone growth as it has a positive effect on the epiphyseal growth plate. SIRT1 is also implicated in various cellular processes, including metabolism, cell cycle regulation, apoptosis, oxidative stress response, and DNA repair. Thus, its expression varies depending on the different metabolic states. During malnutrition, SIRT1 blocks GH signal transduction in hepatocytes to reduce the IGF-1 secretion and prevent hypoglycemia (i.e., it causes transient GH resistance). In this review, we focused on the influence of SIRT1 on GH signal transduction and the implications that may arise for growth processes in children.
Collapse
Affiliation(s)
- Anna Fedorczak
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| | - Andrzej Lewiński
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital-Research Institute, 93-338 Lodz, Poland
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338 Lodz, Poland
| | - Renata Stawerska
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital-Research Institute, 93-338 Lodz, Poland
- Department of Paediatric Endocrinology, Medical University of Lodz, 93-338 Lodz, Poland
| |
Collapse
|
5
|
Liu HY, Lee CH, Hsu CN, Tain YL. Maternal High-Fat Diet Controls Offspring Kidney Health and Disease. Nutrients 2023; 15:2698. [PMID: 37375602 DOI: 10.3390/nu15122698] [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: 05/05/2023] [Revised: 06/04/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
A balanced diet during gestation is critical for fetal development, and excessive intake of saturated fats during gestation and lactation is related to an increased risk of offspring kidney disease. Emerging evidence indicates that a maternal high-fat diet influences kidney health and disease of the offspring via so-called renal programming. This review summarizes preclinical research documenting the connection between a maternal high-fat diet during gestation and lactation and offspring kidney disease, as well as the molecular mechanisms behind renal programming, and early-life interventions to offset adverse programming processes. Animal models indicate that offspring kidney health can be improved via perinatal polyunsaturated fatty acid supplementation, gut microbiota changes, and modulation of nutrient-sensing signals. These findings reinforce the significance of a balanced maternal diet for the kidney health of offspring.
Collapse
Affiliation(s)
- Hsi-Yun Liu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chen-Hao Lee
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
6
|
Promising hepatoprotective effects of lycopene in different liver diseases. Life Sci 2022; 310:121131. [DOI: 10.1016/j.lfs.2022.121131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 11/07/2022]
|
7
|
Atherosclerosis Plaque Reduction by Lycopene Is Mediated by Increased Energy Expenditure through AMPK and PPARα in ApoE KO Mice Fed with a High Fat Diet. Biomolecules 2022; 12:biom12070973. [PMID: 35883529 PMCID: PMC9313394 DOI: 10.3390/biom12070973] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 02/04/2023] Open
Abstract
Lycopene is a carotenoid found in tomatoes that has potent antioxidant activity. The Mediterranean diet is particularly rich in lycopene, which has well-known beneficial effects on cardiovascular health. We tested the effects of lycopene extract in a group of 20 ApoE knockout mice, fed with a high fat western diet for 14 weeks. Starting from week 3 and up to week 14, the mice were randomly divided into two groups that received lycopene (n = 10) by oral suspension every day at the human equivalent dose of 60 mg/day (0.246 mg/mouse/day), or the vehicle solution (n = 10). The lycopene administration reduced triglycerides and cholesterol blood levels starting from week 6 and continuing through to the end of the experiment (p < 0.001). This reduction was mediated by an enhanced liver expression of PPAR-α and AMPK-α and reduced SREBP levels (p < 0.0001). As a histological red-out, the extent of atherosclerotic plaques and the intima−media thickness in the aorta were significantly reduced by lycopene. In this context, lycopene augmented the Nrf-2 positivity staining in the endothelium, thereby confirming that its antioxidant activity was mediated by this nuclear factor. The positive results obtained in this pre-clinical model further support the use of lycopene extracts to reduce atherosclerosis.
Collapse
|
8
|
Liu H, Chen Y, Wang W, Jiang Z, Ma X, Wang F. Comparison of Global Metabolite for Growing Pigs Fed at Metabolizable Energy Requirement for Maintenance. Front Vet Sci 2022; 9:917033. [PMID: 35898548 PMCID: PMC9309205 DOI: 10.3389/fvets.2022.917033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Though the energy requirement for maintenance is an important part of net energy system, little is known of the metabolic characteristics of maintenance energy expenditure. This study was investigated the effect of feeding level at metabolizable energy requirement for maintenance (FLM) on plasma metabolites in growing pigs. Ten barrows (22.5 ± 0.5 kg BW) were kept in metabolism crates and catheterized in the precaval vein during adaptation period. Pigs were fed a corn-soybean meal diet at 782 kJ ME/kg BW0.6·d-1 during d 1 to 8 and then were refeeding at 2,400 kJ ME/kg BW0.6·d-1 on d 9. Plasma samples of each pig were collected by catheter on the morning of d 1, 3, 5, 7, 9, and 10, respectively, for metabolomics testing. Results showed that the concentration of plasma urea nitrogen decreased under FLM (p < 0.01) and increased significantly after refeeding (p < 0.01). The concentration of total cholesterol, high-density lipoprotein, low-density lipoprotein, and albumin in plasma were decreased significantly after refeeding (p < 0.01). Eleven identified compounds were up-regulated and six ones were down-regulated under FLM. In conclusion, the energy metabolism of growing pigs was relatively stable after 4 days of feeding at FLM.
Collapse
Affiliation(s)
- Hu Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yifan Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Wenhui Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhaoning Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Fenglai Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
9
|
The Role of microRNAs in the Mammary Gland Development, Health, and Function of Cattle, Goats, and Sheep. Noncoding RNA 2021; 7:ncrna7040078. [PMID: 34940759 PMCID: PMC8708473 DOI: 10.3390/ncrna7040078] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
Milk is an integral and therefore complex structural element of mammalian nutrition. Therefore, it is simple to conclude that lactation, the process of producing milk, is as complex as the mammary gland, the organ responsible for this biochemical activity. Nutrition, genetics, epigenetics, disease pathogens, climatic conditions, and other environmental variables all impact breast productivity. In the last decade, the number of studies devoted to epigenetics has increased dramatically. Reports are increasingly describing the direct participation of microRNAs (miRNAs), small noncoding RNAs that regulate gene expression post-transcriptionally, in the regulation of mammary gland development and function. This paper presents a summary of the current state of knowledge about the roles of miRNAs in mammary gland development, health, and functions, particularly during lactation. The significance of miRNAs in signaling pathways, cellular proliferation, and the lipid metabolism in agricultural ruminants, which are crucial in light of their role in the nutrition of humans as consumers of dairy products, is discussed.
Collapse
|
10
|
Alzheimer's Disease and Type 2 Diabetes Mellitus: The Use of MCT Oil and a Ketogenic Diet. Int J Mol Sci 2021; 22:ijms222212310. [PMID: 34830192 PMCID: PMC8624628 DOI: 10.3390/ijms222212310] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 12/20/2022] Open
Abstract
Recently, type 2 diabetes mellitus (T2DM) has been reported to be strongly associated with Alzheimer’s disease (AD). This is partly due to insulin resistance in the brain. Insulin signaling and the number of insulin receptors may decline in the brain of T2DM patients, resulting in impaired synaptic formation, neuronal plasticity, and mitochondrial metabolism. In AD patients, hypometabolism of glucose in the brain is observed before the onset of symptoms. Amyloid-β accumulation, a main pathology of AD, also relates to impaired insulin action and glucose metabolism, although ketone metabolism is not affected. Therefore, the shift from glucose metabolism to ketone metabolism may be a reasonable pathway for neuronal protection. To promote ketone metabolism, medium-chain triglyceride (MCT) oil and a ketogenic diet could be introduced as an alternative source of energy in the brain of AD patients.
Collapse
|
11
|
Paulazo MA, Sodero AO. SIRT-1 Activity Sustains Cholesterol Synthesis in the Brain. Neuroscience 2021; 476:116-124. [PMID: 34600072 DOI: 10.1016/j.neuroscience.2021.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 11/18/2022]
Abstract
SIRT-1 is a potent energy regulator that has been implicated in the aging of different tissues, and cholesterol synthesis demands high amounts of cellular adenosine triphosphate. An efficient synaptic transmission depends on processes that are highly influenced by cholesterol levels, like endocytosis, exocytosis and membrane lateral diffusion of neurotransmitter receptors. We set out to investigate whether SIRT-1 activity affects brain cholesterol metabolism. We found that pharmacological inhibition of SIRT-1 with EX-527 reduces the mRNA amounts of 3-hydroxy-3-methylglutaryl-Coenzyme A reductase (HMGCR), Cytochrome P450 46A1 (CYP46A1) and Apolipoprotein E (APO-E) in rat primary cortical cultures. The decreased expression of these genes was paralleled by a significant reduction of the cholesterol levels in this type of neuronal culture. Interestingly, a cholesterol decrease of similar extent was observed in mouse astroglial cultures after EX-527 treatment. In agreement, mice administered with EX-527 for 5 days showed a down-regulation of cholesterol synthesis in the cortex, with significant reductions in the mRNA amounts of the transcription factor Sterol Regulatory Element Binding Protein 2 (SREBP-2) and the enzyme HMGCR, two key regulators of the cholesterol synthesis. These transcriptional changes were paralleled by reduced cholesterol levels at cortical synapses. SIRT-1 inhibition also reduced the amount of cholesterol in the hippocampus but without affecting the HMGCR expression levels. Altogether, these results uncover a role for SIRT-1 in the regulation of cholesterol metabolism, and demonstrate that SIRT-1 is required to sustain adequate levels of cholesterol synthesis in the adult brain.
Collapse
Affiliation(s)
- María A Paulazo
- Institute of Biomedical Research (BIOMED), Pontifical Catholic University of Argentina (UCA) and National Scientific and Technical Research Council (CONICET), C1107AFF Buenos Aires, Argentina
| | - Alejandro O Sodero
- Institute of Biomedical Research (BIOMED), Pontifical Catholic University of Argentina (UCA) and National Scientific and Technical Research Council (CONICET), C1107AFF Buenos Aires, Argentina.
| |
Collapse
|
12
|
Rodrigues LGF, de Araujo LD, Roa SLR, Bueno AC, Uchoa ET, Antunes-Rodrigues J, Moreira AC, Elias LLK, de Castro M, Martins CS. Restricted feeding modulates peripheral clocks and nutrient sensing pathways in rats. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 65:549-561. [PMID: 34591411 PMCID: PMC10528573 DOI: 10.20945/2359-3997000000407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/29/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE Feeding restriction in rats alters the oscillators in suprachiasmatic, paraventricular, and arcuate nuclei, hypothalamic areas involved in food intake. In the present study, using the same animals and experimental protocol, we aimed to analyze if food restriction could reset clock genes (Clock, Bmal1) and genes involved in lipid metabolism (Pgc1a, Pparg, Ucp2) through nutrient-sensing pathways (Sirt1, Ampk, Nampt) in peripheral tissues. METHODS Rats were grouped according to food access: Control group (CG, food ad libitum), Restricted night-fed (RF-n, food access during 2 h at night), Restricted day-fed (RF-d, food access during 2 h in the daytime), and Day-fed (DF, food access during 12 h in the daytime). After 21 days, rats were decapitated at ZT3 (0900-1000 h), ZT11 (1700-1800 h), or ZT17 (2300-2400 h). Blood, liver, brown (BAT) and peri-epididymal (PAT) adipose tissues were collected. Plasma corticosterone and gene expression were evaluated by radioimmunoassay and qPCR, respectively. RESULTS In the liver, the expression pattern of Clock and Bmal1 shifted when food access was dissociated from rat nocturnal activity; this phenomenon was attenuated in adipose tissues. Daytime feeding also inverted the profile of energy-sensing and lipid metabolism-related genes in the liver, whereas calorie restriction induced a pre-feeding increased expression of these genes. In adipose tissues, Sirt1 expression was modified by daytime feeding and calorie restriction, with concomitant expression of Pgc1a, Pparg, and Ucp2 but not Ampk and Nampt. CONCLUSION Feeding restriction reset clock genes and genes involved in lipid metabolism through nutrient-sensing-related genes in rat liver, brown, and peri-epididymal adipose tissues.
Collapse
Affiliation(s)
- Luis Guilherme F Rodrigues
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Leonardo D de Araujo
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Silvia L R Roa
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Ana C Bueno
- Departamento de Pediatria, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Ernane T Uchoa
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - José Antunes-Rodrigues
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Ayrton C Moreira
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Lucila L K Elias
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Margaret de Castro
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Clarissa S Martins
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil,
| |
Collapse
|
13
|
Wu YJ, Fang WJ, Pan S, Zhang SS, Li DF, Wang ZF, Chen WG, Yin Q, Zuo J. Regulation of Sirt1 on energy metabolism and immune response in rheumatoid arthritis. Int Immunopharmacol 2021; 101:108175. [PMID: 34689102 DOI: 10.1016/j.intimp.2021.108175] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/21/2022]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease. Synovial hyperplasia and persistent inflammation serve as its typical pathological manifestations, which ultimately lead to joint destruction and function loss. Both clinical observations and metabolomics studies have revealed the prevalence of metabolic disorders in RA. In inflammatory immune microenvironments, energy metabolism is profoundly changed. Increasingly evidences suggest that this abnormality is involved in the occurrence and development of RA-related inflammation. Unsurprisingly, many energy metabolism sensors have been confirmed with immunoregulatory properties. As a representative, silent information regulator type 1 (Sirt1) controls many aspects of immune cells, such as cell lifespan, polarization, and secretion by functioning as a transcriptional regulator. Because of the profound clinical implication, researches on Sirt1 in the regulation of energy metabolism and immune functions under RA conditions have gradually gained momentum. This signaling balances glycolysis, lipid metabolism and insulin secretion orchestrating with other metabolism sensors, and consequently affects immune milieu through a so-called metabolism-immune feedback mechanism. This article reviews the involvement of Sirt1 in RA by discussing its impacts on energy metabolism and immune functions, and specially highlights the potential of Sirt1-targeting anti-rheumatic regimens. It also provides a theoretical basis for clarifying the mystery about the high incidence of metabolic complications in RA patients and identifying new anti-rheumatic reagents.
Collapse
Affiliation(s)
- Yi-Jin Wu
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, China; Xin'An Medicine Research Center, Wannan Medical College, Wuhu, China
| | - Wen-Juan Fang
- The Second People's Hospital of Hefei, Heifei, China
| | - Shu Pan
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, China; Xin'An Medicine Research Center, Wannan Medical College, Wuhu, China
| | - Sa-Sa Zhang
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, China; Xin'An Medicine Research Center, Wannan Medical College, Wuhu, China
| | - Dan-Feng Li
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Zhong-Fang Wang
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Wen-Gang Chen
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Qin Yin
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, China.
| | - Jian Zuo
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China; Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China.
| |
Collapse
|
14
|
Ingram DK, Pistell PJ, Wang ZQ, Yu Y, Massimino S, Davenport GM, Hayek M, Roth GS. Characterization and Mechanisms of Action of Avocado Extract Enriched in Mannoheptulose as a Candidate Calorie Restriction Mimetic. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7367-7376. [PMID: 34170139 DOI: 10.1021/acs.jafc.1c01995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Increased consumer interest in the avocado (Persea americana or Persea gratissima) has been attributed to established health benefits of this fruit associated with a wide range of ingredients. In search of effective calorie restriction mimetics (CRM), we present herein a consideration of possible health benefits of the rare sugar, mannoheptulose (MH), which acts as an intracellular glycolytic inhibitor and presents the highest concentration of this inhibitor in unripe avocados. A method for producing an extract of unripe avocado (AvX) to enrich concentrations of MH is described. Experiments using myocyte cultures demonstrated a pattern of CRM-like responses when treated with AvX. In vivo experiments confirmed that orally consumed AvX is bioavailable in both mice and dogs, as observed in urine and blood samples. Additional experiments in both these species demonstrated CRM-like improvements in glucose and insulin responses. In sum, the MH-enriched AvX exhibits promise as a CRM.
Collapse
Affiliation(s)
- Donald K Ingram
- Pennington Biomedical Research Center, Louisiana State University, 5600 Perkins Road, Baton Rouge, Louisiana 70808, United States
| | - Paul J Pistell
- Department of Psychology, Towson University, 8000 York Road, Towson, Maryland 21252, United States
| | - Zhong Q Wang
- Pennington Biomedical Research Center, Louisiana State University, 5600 Perkins Road, Baton Rouge, Louisiana 70808, United States
| | - Yongmei Yu
- Pennington Biomedical Research Center, Louisiana State University, 5600 Perkins Road, Baton Rouge, Louisiana 70808, United States
| | - Stefan Massimino
- Kaiser Permanente Center for Health Research, 3800 N Interstate Ave, Portland, Oregon 97227, United States
| | - Gary M Davenport
- Archer Daniels Midland Company, 115 W National Road, P.O. Box 310, Englewood, Ohio 46322, United States
| | - Michael Hayek
- Companion Animal Technical Services, Land O'Lakes, 4001 Lexington Ave N, Arden Hills, Minnesota 55126, United States
| | - George S Roth
- GeroScience, Inc, 1124 Ridge Road, Pylesville, Maryland 21132, United States
| |
Collapse
|
15
|
Duque G, Al Saedi A, Rivas D, Miard S, Ferland G, Picard F, Gaudreau P. Differential Effects of Long-Term Caloric Restriction and Dietary Protein Source on Bone and Marrow Fat of the Aging Rat. J Gerontol A Biol Sci Med Sci 2021; 75:2031-2036. [PMID: 32298404 DOI: 10.1093/gerona/glaa093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Indexed: 01/11/2023] Open
Abstract
Long-term caloric restriction (CR) has been shown to be beneficial to various tissues and organs. In contrast, CR exerts differential effects on bone, which could be due in part to the nature of the protein regime utilized. Male Sprague Dawley rats (8-month-old) were subjected for 12 months to 40% CR in macronutrients and compared with rats fed ad libitum for the same period. Casein- and soy-fed groups were compared. There was a significant decrease in bone quality in both CR groups, which was independent of the source of protein in the diet. In contrast, the group fed soy protein ad libitum showed better bone quality and higher levels of bone formation compared with casein-fed animals. Notably, bone marrow adipocytes were not mobilized upon CR as demonstrated by an absence of change in adipocyte number and tissue expression of leptin. This study demonstrates that the negative effect of CR on bone quality could not be prevented by the most common protein regimes.
Collapse
Affiliation(s)
- Gustavo Duque
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, Australia.,Department of Medicine-Western Health, University of Melbourne, St. Albans, Australia
| | - Ahmed Al Saedi
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, Australia.,Department of Medicine-Western Health, University of Melbourne, St. Albans, Australia
| | - Daniel Rivas
- Lady Davis Institute for Medical Research, Montreal, Canada
| | | | - Guylaine Ferland
- Département de nutrition, Faculté de médecine de l'Université de Montréal (UdeM), Canada.,Centre de recherche de l'Institut de cardiologie de Montréal, Canada
| | - Frederic Picard
- Quebec Heart and Lung Institute, Canada.,Faculty of Pharmacy, Université Laval, Québec, Canada
| | - Pierrette Gaudreau
- Département de médecine, UdeM, Montreal, Canada.,Laboratoire de Neuroendocrinologie du vieillissement, Centre de Recherche du Centre hospitalier de l'UdeM, Montréal, Canada
| |
Collapse
|
16
|
Khanahmadi M, Manafi B, Tayebinia H, Karimi J, Khodadadi I. Downregulation of Sirt1 is correlated to upregulation of p53 and increased apoptosis in epicardial adipose tissue of patients with coronary artery disease. EXCLI JOURNAL 2020; 19:1387-1398. [PMID: 33250679 PMCID: PMC7689241 DOI: 10.17179/excli2020-2423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/01/2020] [Indexed: 12/31/2022]
Abstract
The higher expression level of p53 in epithelial adipose tissue (EAT) has previously been reported in atherosclerosis. Since we hypothesized that the expression of p53 is modulated by Sirt1, the aim of this study was to determine the expression levels of Sirt1 and p53 and to investigate their correlation to apoptosis in EAT of patients with coronary artery disease (CAD). Thirty-five patients with more than 50 % stenosis in at least one of the main coronary arteries were considered as CAD group while 29 patients with no clinical signs of atherosclerosis who underwent open-heart surgery for valve replacement were classified as control group. EAT biopsy samples were collected from all participants during surgery. Sirt1, p53, Bax, and Bcl-2 gene expression levels were determined in EAT by qRT-PCR and Western blotting was carried out to assess Sirt1 and p53 protein levels. Hematoxylin and eosin staining was used for histopathological analysis. mRNA and protein levels of Sirt1 in EAT were significantly lower in patients with CAD compared with control group, whereas CAD patients showed greater p53 gene and protein expressions. In addition, inverse correlations were observed between Sirt1 and p53 at both mRNA and protein levels. The Bax and ratio of Bax/Bcl-2 gene expressions were higher in CAD group, but no difference was observed in Bcl-2 expression. Histopathological analysis showed apoptotic bodies and infiltrated immune cells in EAT of CAD group. Our results suggest that the Sirt1-p53 axis may involve in atherosclerosis by promotion of apoptosis.
Collapse
Affiliation(s)
- Mahdieh Khanahmadi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Babak Manafi
- Department of Surgery, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Heidar Tayebinia
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
17
|
García-García FJ, Monistrol-Mula A, Cardellach F, Garrabou G. Nutrition, Bioenergetics, and Metabolic Syndrome. Nutrients 2020; 12:E2785. [PMID: 32933003 PMCID: PMC7551996 DOI: 10.3390/nu12092785] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
According to the World Health Organization (WHO), the global nutrition report shows that whilst part of the world's population starves, the other part suffers from obesity and associated complications. A balanced diet counterparts these extreme conditions with the proper proportion, composition, quantity, and presence of macronutrients, micronutrients, and bioactive compounds. However, little is known on the way these components exert any influence on our health. These nutrients aiming to feed our bodies, our tissues, and our cells, first need to reach mitochondria, where they are decomposed into CO2 and H2O to obtain energy. Mitochondria are the powerhouse of the cell and mainly responsible for nutrients metabolism, but they are also the main source of oxidative stress and cell death by apoptosis. Unappropriated nutrients may support mitochondrial to become the Trojan horse in the cell. This review aims to provide an approach to the role that some nutrients exert on mitochondria as a major contributor to high prevalent Western conditions including metabolic syndrome (MetS), a constellation of pathologic conditions which promotes type II diabetes and cardiovascular risk. Clinical and experimental data extracted from in vitro animal and cell models further demonstrated in patients, support the idea that a balanced diet, in a healthy lifestyle context, promotes proper bioenergetic and mitochondrial function, becoming the best medicine to prevent the onset and progression of MetS. Any advance in the prevention and management of these prevalent complications help to face these challenging global health problems, by ameliorating the quality of life of patients and reducing the associated sociosanitary burden.
Collapse
Affiliation(s)
- Francesc Josep García-García
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Anna Monistrol-Mula
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Francesc Cardellach
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Glòria Garrabou
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| |
Collapse
|
18
|
Fahlbusch P, Knebel B, Hörbelt T, Barbosa DM, Nikolic A, Jacob S, Al-Hasani H, Van de Velde F, Van Nieuwenhove Y, Müller-Wieland D, Lapauw B, Ouwens DM, Kotzka J. Physiological Disturbance in Fatty Liver Energy Metabolism Converges on IGFBP2 Abundance and Regulation in Mice and Men. Int J Mol Sci 2020; 21:ijms21114144. [PMID: 32532003 PMCID: PMC7312731 DOI: 10.3390/ijms21114144] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022] Open
Abstract
Fatty liver occurs from simple steatosis with accumulated hepatic lipids and hepatic insulin resistance to severe steatohepatitis, with aggravated lipid accumulation and systemic insulin resistance, but this progression is still poorly understood. Analyses of hepatic gene expression patterns from alb-SREBP-1c mice with moderate, or aP2-SREBP-1c mice with aggravated, hepatic lipid accumulation revealed IGFBP2 as key nodal molecule differing between moderate and aggravated fatty liver. Reduced IGFBP2 expression in aggravated fatty liver was paralleled with promoter hypermethylation, reduced hepatic IGFBP2 secretion and IGFBP2 circulating in plasma. Physiologically, the decrease of IGFBP2 was accompanied with reduced fatty acid oxidation and increased de novo lipogenesis potentially mediated by IGF1 in primary hepatocytes. Furthermore, methyltransferase and sirtuin activities were enhanced. In humans, IGFBP2 serum concentration was lower in obese men with non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH) compared to non-obese controls, and liver fat reduction by weight-loss intervention correlated with an increase of IGFBP2 serum levels. In conclusion, hepatic IGFBP2 abundance correlates to its circulating level and is related to hepatic energy metabolism and de novo lipogenesis. This designates IGFBP2 as non-invasive biomarker for fatty liver disease progression and might further provide an additional variable for risk prediction for pathogenesis of fatty liver in diabetes subtype clusters.
Collapse
Affiliation(s)
- Pia Fahlbusch
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Birgit Knebel
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Tina Hörbelt
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - David Monteiro Barbosa
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Aleksandra Nikolic
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Sylvia Jacob
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Hadi Al-Hasani
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Frederique Van de Velde
- Department of Endocrinology, Ghent University Hospital, 9000 Ghent, Belgium; (F.V.d.V.); (B.L.)
| | - Yves Van Nieuwenhove
- Department of Gastrointestinal Surgery, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Dirk Müller-Wieland
- Clinical Research Centre, Department of Internal Medicine I, University Hospital Aachen, 52074 Aachen, Germany;
| | - Bruno Lapauw
- Department of Endocrinology, Ghent University Hospital, 9000 Ghent, Belgium; (F.V.d.V.); (B.L.)
| | - D. Margriet Ouwens
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
- Department of Endocrinology, Ghent University Hospital, 9000 Ghent, Belgium; (F.V.d.V.); (B.L.)
| | - Jorg Kotzka
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany; (P.F.); (B.K.); (T.H.); (D.M.B.); (A.N.); (S.J.); (H.A.-H.); (D.M.O.)
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
- Correspondence: ; Tel.: +49-211-3382-537
| |
Collapse
|
19
|
Simó-Mirabet P, Perera E, Calduch-Giner JA, Pérez-Sánchez J. Local DNA methylation helps to regulate muscle sirtuin 1 gene expression across seasons and advancing age in gilthead sea bream ( Sparus aurata). Front Zool 2020; 17:15. [PMID: 32467713 PMCID: PMC7227224 DOI: 10.1186/s12983-020-00361-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/30/2020] [Indexed: 12/15/2022] Open
Abstract
Background Sirtuins (SIRTs) are master regulators of metabolism, and their expression patterns in gilthead sea bream (GSB) reveal different tissue metabolic capabilities and changes in energy status. Since little is known about their transcriptional regulation, the aim of this work was to study for the first time in fish the effect of age and season on sirt gene expression, correlating expression patterns with local changes in DNA methylation in liver and white skeletal muscle (WSM). Methods Gene organization of the seven sirts was analyzed by BLAT searches in the IATS-CSIC genomic database (www.nutrigroup-iats.org/seabreamdb/). The presence of CpG islands (CGIs) was mapped by means of MethPrimer software. DNA methylation analyses were performed by bisulfite pyrosequencing. A PCR array was designed for the simultaneous gene expression profiling of sirts and related markers (cs, cpt1a, pgc1α, ucp1, and ucp3) in the liver and WSM of one- and three-year-old fish during winter and summer. Results The occurrence of CGIs was evidenced in the sirt1 and sirt3 promoters. This latter CGI remained hypomethylated regardless of tissue, age and season. Conversely, DNA methylation of sirt1 at certain CpG positions within the promoter varied with age and season in the WSM. Among them, changes at several SP1 binding sites were negatively correlated with the decrease in sirt1 expression in summer and in younger fish. Changes in sirt1 regulation match well with variations in feed intake and energy metabolism, as judged by the concurrent changes in the analyzed markers. This was supported by discriminant analyses, which identified sirt1 as a highly responsive element to age- and season-mediated changes in energy metabolism in WSM. Conclusions The gene organization of SIRTs is highly conserved in vertebrates. GSB sirt family members have CGI- and non-CGI promoters, and the presence of CGIs at the sirt1 promoter agrees with its ubiquitous expression. Gene expression analyses support that sirts, especially sirt1, are reliable markers of age- and season-dependent changes in energy metabolism. Correlation analyses suggest the involvement of DNA methylation in the regulation of sirt1 expression, but the low methylation levels suggest the contribution of other putative mechanisms in the transcriptional regulation of sirt1.
Collapse
Affiliation(s)
- Paula Simó-Mirabet
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain
| | - Erick Perera
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain
| | - Josep Alvar Calduch-Giner
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain
| |
Collapse
|
20
|
Abstract
In this research paper we filter and verify miRNAs which may target silent information regulator homolog 2 (SIRT2) gene and then describe the mechanism whereby miRNA-212 might regulate lipogenic genes in mammary epithelial cell lines via targeting SIRT2. Bioinformatics analysis revealed that the bovine SIRT2 gene is regulated by three miRNAs: miR-212, miR-375 and miR-655. The three miRNAs were verified and screened by qRT-PCR, western blot, and luciferase multiplex verification techniques and only miR-212 was shown to have a targeting relationship with SIRT2. The results of co-transfecting miR-212 and silencing RNA (siRNA) showed that by targeting SIRT2, miR-212 can regulate the expression of fatty acid synthetase (FASN) and sterol regulatory element binding factor 1 (SREBP1) but not peroxisome proliferator-activated receptor gamma (PPARγ). Measurement of triglyceride (TAG) content showed that miR-212 increased the fat content of mammary epithelial cell lines. The study indicates that miR-212 could target and inhibit the expression of the SIRT2 gene to promote lipogenesis in mammary epithelial cell lines.
Collapse
|
21
|
Fatty liver diseases, mechanisms, and potential therapeutic plant medicines. Chin J Nat Med 2020; 18:161-168. [PMID: 32245585 DOI: 10.1016/s1875-5364(20)30017-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Indexed: 02/07/2023]
Abstract
The liver is an important metabolic organ and controls lipid, glucose and energy metabolism. Dysruption of hepatic lipid metabolism is often associated with fatty liver diseases, including nonalcoholic fatty liver disease (NAFLD), alcoholic fatty liver diseases (AFLD) and hyperlipidemia. Recent studies have uncovered the contribution of hormones, transcription factors, and inflammatory cytokines to the pathogenesis of dyslipidemia and fatty liver diseases. Moreover, a significant amount of effort has been put to examine the mechanisms underlying the potential therapeutic effects of many natural plant products on fatty liver diseases and metabolic diseases. We review the current understanding of insulin, thyroid hormone and inflammatory cytokines in regulating hepatic lipid metabolism, focusing on several essential transcription regulators, such as Sirtuins (SIRTs), Forkhead box O (FoxO), Sterol-regulatory element-binding proteins (SREBPs). We also discuss a few representative natural products with promising thereapeutic effects on fatty liver disease and dyslipidemia.
Collapse
|
22
|
Tagliari CFDS, de Oliveira CN, Vogel GM, da Silva PB, Linden R, Lazzaretti RK, Notti RK, Sprinz E, Mattevi VS. Investigation of SIRT1 gene variants in HIV-associated lipodystrophy and metabolic syndrome. Genet Mol Biol 2020; 43:e20190142. [PMID: 32106282 PMCID: PMC7198015 DOI: 10.1590/1678-4685-gmb-2019-0142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 11/21/2019] [Indexed: 01/17/2023] Open
Abstract
HIV-infected individuals on chronic use of highly active antiretroviral therapy (HAART) are more likely to develop adipose tissue and metabolic disorders, such as lipodystrophy (LD) and metabolic syndrome (MetS). The development of these phenotypes is known to be multifactorial. Thus, variants in genes implicated in adipogenesis and lipid metabolism may increase susceptibility to LD and MetS. Sirtuin 1 (SIRT1) may influence the outcome of these disturbances due to its role in the regulation of transcription factors involved in energy regulation. Therefore, we genotyped four polymorphisms located in SIRT1 (rs2273773 T>C, rs12413112 G>A, rs7895833 A>G, rs12049646 T>C) in 832 HIV-infected patients receiving HAART by real-time polymerase chain reaction. The prevalence of LD was 55.8% and MetS was 35.3%. Lipoatrophy was the most prevalent subtype in all samples (38.0%) and showed significant difference between white and non-white individuals (P = 0.002). None of the genetic variants investigated in SIRT1 was associated with LD and MetS. White individuals and those in longer time of HAART use were more likely to develop LD. We concluded that these SIRT1 polymorphisms are not predictive factors to the development of lipodystrophy and metabolic syndrome in HIV-infected individuals from Brazil.
Collapse
Affiliation(s)
| | - Cáren Nunes de Oliveira
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Laboratório de Biologia Molecular, Porto Alegre, RS, Brazil
| | - Greice Meyer Vogel
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Laboratório de Biologia Molecular, Porto Alegre, RS, Brazil
| | - Patrícia Baptista da Silva
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Laboratório de Biologia Molecular, Porto Alegre, RS, Brazil
| | - Rafael Linden
- Universidade Feevale, Instituto de Ciências da Saúde, Laboratório de Toxicologia Analítica, Novo Hamburgo, RS, Brazil
| | - Rosmeri Kuhmmer Lazzaretti
- Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Departamento de Doenças Infecciosas, Porto Alegre, RS, Brazil
| | - Regina Kuhmmer Notti
- Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Departamento de Doenças Infecciosas, Porto Alegre, RS, Brazil
| | - Eduardo Sprinz
- Universidade Federal do Rio Grande do Sul, Hospital de Clínicas de Porto Alegre, Departamento de Doenças Infecciosas, Porto Alegre, RS, Brazil
| | - Vanessa Suñé Mattevi
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Laboratório de Biologia Molecular, Porto Alegre, RS, Brazil
| |
Collapse
|
23
|
Yu J, Li J, He S, Xu L, Zhang Y, Jiang H, Gong D, Gu Z. Sirt1 regulates the expression of critical metabolic genes in chicken hepatocytes. ANIMAL PRODUCTION SCIENCE 2020. [DOI: 10.1071/an18606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Context
Studies in mammals show that SIRT1 plays an important role in many biological processes including liver metabolism through histone and non-histone deacetylation. Little is known about the function of Sirt1 in the chicken.
Aims
The current study investigated the expression pattern of Sirt1 mRNA in the chicken and its functions in the chicken liver.
Methods
In this work, we used real-time quantitative polymerase chain reaction to quantify the expression levels of Sirt1 mRNA in major chicken organs and tissue types, siRNA to knock down Sirt1 expression in primary chicken hepatocytes, RNA sequencing to identify gene-expression changes induced by Sirt1 knockdown, and analysed the function of the differentially expressed genes (DEGs) through gene ontology enrichment and Kyoto Encyclopedia of Genes and Genomes ontology analysis.
Key results
In total, 86 DEGs were found between Sirt1 knockdown and control chicken hepatocytes, of which 63 genes were downregulated and 23 genes were upregulated by Sirt1 knockdown. The Kyoto Encyclopedia of Genes and Genomes analysis showed that 24 DEGs were involved in metabolism. Seven DEGs were involved in carbohydrate and lipid metabolism.
Conclusions
The present study showed that Sirt1 regulates the expression of genes involved in carbohydrate and lipid metabolism and many other biological processes in the chicken liver.
Implications
The results of the present study imply that Sirt1 has various functions in the chicken liver and that Sirt1 plays a potentially important role in hepatic carbohydrate and lipid metabolism in the chicken.
Collapse
|
24
|
Dikalov SI, Dikalova AE. Crosstalk Between Mitochondrial Hyperacetylation and Oxidative Stress in Vascular Dysfunction and Hypertension. Antioxid Redox Signal 2019; 31:710-721. [PMID: 30618267 PMCID: PMC6708267 DOI: 10.1089/ars.2018.7632] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: Vascular dysfunction plays a key role in the development of arteriosclerosis, heart disease, and hypertension, which causes one-third of deaths worldwide. Vascular oxidative stress and metabolic disorders contribute to vascular dysfunction, leading to impaired vasorelaxation, vascular hypertrophy, fibrosis, and aortic stiffening. Mitochondria are critical in the regulation of metabolic and antioxidant functions; therefore, mitochondria-targeted treatments could be beneficial. Recent Advances: Vascular dysfunction is crucial in hypertension pathophysiology and exhibits bidirectional relationship. Metabolic disorders and oxidative stress contribute to the pathogenesis of vascular dysfunction and hypertension, which are associated with mitochondrial impairment and hyperacetylation. Mitochondrial deacetylase Sirtuin 3 (Sirt3) is critical in the regulation of metabolic and antioxidant functions. Clinical studies show that cardiovascular disease risk factors reduce Sirt3 level and Sirt3 declines with age, paralleling the increased incidence of cardiovascular disease and hypertension. An imbalance between mitochondrial acetylation and reduced Sirt3 activity contributes to mitochondrial dysfunction and oxidative stress. We propose that mitochondrial hyperacetylation drives a vicious cycle between metabolic disorders and mitochondrial oxidative stress, promoting vascular dysfunction and hypertension. Critical Issues: The mechanisms of mitochondrial dysfunction are still obscure in human hypertension. Mitochondrial hyperacetylation and oxidative stress contribute to mitochondrial dysfunction; however, regulation of mitochondrial acetylation, the role of GCN5L1 (acetyl-CoA-binding protein promoting acetyltransferase protein acetylation) acetyltransferase, Sirt3 deacetylase, and acetylation of specific proteins require further investigations. Future Directions: There is an urgent need to define molecular mechanisms and the pathophysiological role of mitochondrial hyperacetylation, identify novel pharmacological targets, and develop therapeutic approaches to reduce this phenomenon.
Collapse
Affiliation(s)
- Sergey I Dikalov
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna E Dikalova
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
25
|
Kim JW, Lee YJ, You YH, Moon MK, Yoon KH, Ahn YB, Ko SH. Effect of sodium-glucose cotransporter 2 inhibitor, empagliflozin, and α-glucosidase inhibitor, voglibose, on hepatic steatosis in an animal model of type 2 diabetes. J Cell Biochem 2019; 120:8534-8546. [PMID: 30474134 DOI: 10.1002/jcb.28141] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 11/05/2018] [Indexed: 01/24/2023]
Abstract
OBJECTIVE We investigated the effects of sodium-glucose cotransporter 2 inhibitor, empagliflozin, and α-glucosidase inhibitor, voglibose, on hepatic steatosis in an animal model of type 2 diabetes (T2DM). METHODS Empagliflozin (OLETF-EMPA) or voglibose (OLETF-VOG) was administered to Otsuka Long-Evans Tokushima fatty (OLETF) rats once daily for 12 weeks. Control Long-Evans Tokushima Otsuka (LETO) and OLETF (OLETF-C) rats received saline. RESULTS Blood glucose levels were significantly suppressed in OLETF-EMPA and OLETF-VOG compared with the OLETF-C group. The liver fat content was significantly higher in the OLETF-C group than in the OLETF-EMPA and OLETF-VOG. Hepatic gene expressions involved in gluconeogenesis (glucose 6-phosphatase [G6Pase], fructose-1,6-bisphosphatase [FBP1], and phosphoenolpyruvate carboxykinase [PEPCK]) and lipogenesis (acetyl-CoA carboxylase [ACC], fatty acid synthase [FAS], and sterol regulatory element-binding transcription factor 1c [SREBP-1c]) were significantly decreased in the OLETF-EMPA group compared with other OLETF groups (OLETF-C and OLETF-VOG). Sirtuin 1 (SIRT1) expression level and SIRT1 activity were markedly reduced in OLETF-C rats; however, its expression increased in the OLETF-EMPA and OLETF-VOG. AMP-activated protein kinase (AMPK) phosphorylation level was remarkably increased by empagliflozin treatment in OLETF rats compared with other OLETF groups. Long-term empagliflozin and voglibose treatment reduced hepatic steatosis with suppression of gluconeogenesis and lipogenesis pathway in OLETF rats. CONCLUSION We suggest that this metabolic improvement might be related to SIRT1 and AMPK pathway in T2DM. But empagliflozin is thought to have more advantage to prevent hepatic steatosis than voglibose in T2DM.
Collapse
Affiliation(s)
- Ji-Won Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ye-Jee Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Young-Hye You
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Kyong Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul University College of Medicine, Seoul, Korea
| | - Kun-Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yu-Bae Ahn
- Division of Endocrinology and Metabolism, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Hyun Ko
- Division of Endocrinology and Metabolism, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
26
|
Camptothecin activates SIRT1 to promote lipid catabolism through AMPK/FoxO1/ATGL pathway in C2C12 myogenic cells. Arch Pharm Res 2019; 42:672-683. [DOI: 10.1007/s12272-019-01155-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 04/15/2019] [Indexed: 01/02/2023]
|
27
|
Liu H, Chen Y, Li Z, Li Y, Lai C, Piao X, van Milgen J, Wang F. Metabolizable energy requirement for maintenance estimated by regression analysis of body weight gain or metabolizable energy intake in growing pigs. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 32:1397-1406. [PMID: 30744343 PMCID: PMC6722305 DOI: 10.5713/ajas.17.0898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/08/2019] [Indexed: 01/28/2023]
Abstract
Objective Feed energy required for pigs is first prioritized to meet maintenance costs. Additional energy intake in excess of the energy requirement for maintenance is retained as protein and fat in the body, leading to weight gain. The objective of this study was to estimate the metabolizable energy requirements for maintenance (MEm) by regressing body weight (BW) gain against metabolizable energy intake (MEI) in growing pigs. Methods Thirty-six growing pigs (26.3±1.7 kg) were allotted to 1 of 6 treatments with 6 replicates per treatment in a randomized complete block design. Treatments were 6 feeding levels which were calculated as 50%, 60%, 70%, 80%, 90%, or 100% of the estimated ad libitum MEI (2,400 kJ/kg BW0.60 d). All pigs were individually housed in metabolism crates for 30 d and weighed every 5 d. Moreover, each pig from each treatment was placed in the open-circuit respiration chambers to measure heat production (HP) and energy retained as protein (REp) and fat (REf) every 5 d. Serum biochemical parameters of pigs were analyzed at the end of the experiment. Results The average daily gain (ADG) and HP as well as the REp and REf linearly increased with increasing feed intake (p<0.010). β-hydroxybutyrate concentration of serum tended to increase with increasing feed intake (p = 0.080). The regression equations of MEI on ADG were MEI, kJ/kg BW0.60 d = 1.88×ADG, g/d+782 (R2 = 0.86) and MEm was estimated at 782 kJ/kg BW0.60 d. Protein retention of growing pigs would be positive while REf would be negative at this feeding level via regression equations of REp and REf on MEI. Conclusion The MEm was estimated at 782 kJ/kg BW0.60 d in current experiment. Furthermore, growing pigs will deposit protein and oxidize fat if provided feed at the estimated maintenance level.
Collapse
Affiliation(s)
- Hu Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yifan Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhongchao Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yakui Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Changhua Lai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiangshu Piao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | | | - Fenglai Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
28
|
Liu H, Li T, Jiang Z, Wang W, Ming D, Chen Y, Wang F. Effect of different time intervals after feeding on plasma metabolites in growing pigs: an UPLC-MS-based metabolomics study. Anim Sci J 2019; 90:554-562. [PMID: 30714268 DOI: 10.1111/asj.13178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/27/2018] [Accepted: 12/11/2018] [Indexed: 11/28/2022]
Abstract
A diet consumed by pigs provides the nutrients for the production of a large number of metabolites that, after first-pass metabolism in the liver, circulate systemically where they may exert diverse physiologic influences on pigs. So far, little is known of how feeding elicits changes in metabolic profiles for growing pigs. This study investigated differences in plasma metabolites in growing pigs at several intervals after feeding using the technique of metabolomics. Ten barrows (22.5 ± 0.5 kg BW) were fed a corn-soybean meal basal diet and were kept in metabolism crates for a period of 11 days. An indwelling catheter was inserted into the jugular vein of each pig before the experimental period. Plasmas before and 1, 4, and 8 hr after feeding were collected at day 11 and differential metabolites were determined using a metabolomics approach. Direct comparison at several intervals after feeding revealed differences in 14 compounds. Identified signatures were enriched in metabolic pathways related to linoleic acid metabolism, arginine and proline metabolism, lysine degradation, glycine, serine and threonine metabolism, and lysine biosynthesis. These results suggest that plasma metabolites of growing pigs after feeding were modulated through changes in linoleic acid metabolism and amino acid metabolism.
Collapse
Affiliation(s)
- Hu Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Tiantian Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Zhaoning Jiang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Wenhui Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Dongxu Ming
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Yifan Chen
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Fenglai Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| |
Collapse
|
29
|
Martínez-Jiménez V, Cortez-Espinosa N, Rodríguez-Varela E, Vega-Cárdenas M, Briones-Espinoza M, Ruíz-Rodríguez VM, López-López N, Briseño-Medina A, Turiján-Espinoza E, Portales-Pérez DP. Altered levels of sirtuin genes (SIRT1, SIRT2, SIRT3 and SIRT6) and their target genes in adipose tissue from individual with obesity. Diabetes Metab Syndr 2019; 13:582-589. [PMID: 30641770 DOI: 10.1016/j.dsx.2018.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Sirtuins regulate energy metabolism and insulin sensitivity through their ability to act as energy sensors and regulators in several metabolic tissues. AIM To evaluate the expression levels of sirtuin genes SIRT1, SIRT2, SIRT3 and SIRT6 and their target genes (PPAR-α, PGC1-α, NRF1, DGAT1, PPAR-γ and FOXO3a) in subcutaneous adipose tissue collected from individuals with normoweight, overweight and obesity. METHODS Adipose tissue samples, obtained by lipoaspiration during liposuction surgery, were processed to obtain RNA, which was reverse-transcribed to cDNA. Then, we measured the expression levels of each gene by qPCR. RESULTS We found differences in the mRNA expression of SIRT1, SIRT2, SIRT3 and SIRT6 and their target genes (PPAR-α, PGC1-α, NRF1, DGAT1, PPAR-γ and FOXO3a) in adipose tissue from overweight or obese subjects when compared to normoweight subjects. All genes analyzed, except SIRT2, showed correlation with BMI. CONCLUSIONS Our findings in human subcutaneous adipose tissue show that increased body mass index modifies the expression of genes encoding sirtuins and their target genes, which are metabolic regulators of adipose tissue. Therefore, these could be used as biomarkers to predict the ability of adipose tissue to gain mass of adipose tissue.
Collapse
Affiliation(s)
- Verónica Martínez-Jiménez
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Nancy Cortez-Espinosa
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Eduardo Rodríguez-Varela
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Mariela Vega-Cárdenas
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Margarita Briones-Espinoza
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Víctor M Ruíz-Rodríguez
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Nallely López-López
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Armando Briseño-Medina
- Aesthetic and Corrective Plastic Surgery Clinic, San Luis Potosí, San Luis Potosi, Mexico
| | - Eneida Turiján-Espinoza
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - Diana P Portales-Pérez
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico.
| |
Collapse
|
30
|
Rickert E, Fernandez MO, Choi I, Gorman M, Olefsky JM, Webster NJG. Neuronal SIRT1 Regulates Metabolic and Reproductive Function and the Response to Caloric Restriction. J Endocr Soc 2018; 3:427-445. [PMID: 30746504 PMCID: PMC6364627 DOI: 10.1210/js.2018-00318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/19/2018] [Indexed: 01/06/2023] Open
Abstract
Sirt1 is an NAD-dependent, class III deacetylase that functions as a cellular energy sensor. In addition to its well-characterized effects in peripheral tissues, emerging evidence suggests that neuronal Sirt1 activity plays a role in the central regulation of energy balance and glucose metabolism. In this study, we generated mice expressing an enzymatically inactive form (N-MUT) or wild-type (WT) SIRT1 (N-OX) in mature neurons. N-OX male and female mice had impaired glucose tolerance, and N-MUT female, but not male, mice had improved glucose tolerance compared with that of WT littermates. Furthermore, glucose tolerance was improved in all mice with caloric restriction (CR) but was greater in the N-OX mice, who had better glucose tolerance than their littermates. At the reproductive level, N-OX females had impaired estrous cycles, with increased cycle length and more time in estrus. LH and progesterone surges were absent on the evening of proestrus in the N-OX mice, suggesting a defect in spontaneous ovulation, which was confirmed by the ovarian histology revealing fewer corpora lutea. Despite this defect, the mice were still fertile when mated to WT mice on the day of proestrus, indicating that the mice could respond to normal pheromonal or environmental cues. When subjected to CR, the N-OX mice went into diestrus arrest earlier than their littermates. Together, these results suggested that the overexpression of SIRT1 rendered the mice more sensitive to the metabolic improvements and suppression of reproductive cycles by CR, which was independent of circadian rhythms.
Collapse
Affiliation(s)
- Emily Rickert
- VA San Diego Healthcare System, San Diego, California.,Department of Medicine, University of California San Diego, La Jolla, California
| | | | - Irene Choi
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Michael Gorman
- Department of Psychology, University of California San Diego, La Jolla, California
| | - Jerrold M Olefsky
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Nicholas J G Webster
- VA San Diego Healthcare System, San Diego, California.,Department of Medicine, University of California San Diego, La Jolla, California.,Moores Cancer Center, University of California San Diego, La Jolla, California
| |
Collapse
|
31
|
α-Mangostin Alleviated Lipopolysaccharide Induced Acute Lung Injury in Rats by Suppressing NAMPT/NAD Controlled Inflammatory Reactions. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5470187. [PMID: 30405740 PMCID: PMC6199890 DOI: 10.1155/2018/5470187] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/08/2018] [Accepted: 09/23/2018] [Indexed: 12/18/2022]
Abstract
α-Mangostin (MAN) is a bioactive xanthone isolated from mangosteen. This study was designed to investigate its therapeutic effects on acute lung injury (ALI) and explore the underlying mechanisms of action. Rats from treatment groups were subject to oral administration of MAN for 3 consecutive days beforehand, and then ALI was induced in all the rats except for normal controls via an intraperitoneal injection with lipopolysaccharide. The severity of disease was evaluated by histological examination and hematological analysis. Protein expressions in tissues and cells were examined with immunohistochemical and immunoblotting methods, respectively. The levels of cytokines and nicotinamide adenine dinucleotide (NAD) were determined using ELISA and colorimetric kits, respectively. It was found that MAN treatment significantly improved histological conditions, reduced leucocytes counts, relieved oxidative stress, and declined TNF-α levels in ALI rats. Meanwhile, MAN treatment decreased expressions of nicotinamide phosphoribosyltransferase (NAMPT) and Sirt1 both in vivo and in vitro, which was accompanied with a synchronized decline of NAD and TNF-α. Immunoblotting assay further showed that MAN downregulated HMGB1, TLR4, and p-p65 in RAW 264.7 cells. MAN induced declines of both HMGB1/TLR4/p-p65 and TNF-α were substantially reversed by cotreatment with nicotinamide mononucleotide or NAD. These results suggest that downregulation of NAMPT/NAD by MAN treatments contributes to the alleviation of TLR4/NF-κB-mediated inflammations in macrophage, which is essential for amelioration of ALI in rats.
Collapse
|
32
|
Hasan-Olive MM, Lauritzen KH, Ali M, Rasmussen LJ, Storm-Mathisen J, Bergersen LH. A Ketogenic Diet Improves Mitochondrial Biogenesis and Bioenergetics via the PGC1α-SIRT3-UCP2 Axis. Neurochem Res 2018; 44:22-37. [PMID: 30027365 DOI: 10.1007/s11064-018-2588-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 11/30/2022]
Abstract
A ketogenic diet (KD; high-fat, low-carbohydrate) can benefit refractory epilepsy, but underlying mechanisms are unknown. We used mice inducibly expressing a mutated form of the mitochondrial DNA repair enzyme UNG1 (mutUNG1) to cause progressive mitochondrial dysfunction selectively in forebrain neurons. We examined the levels of mRNAs and proteins crucial for mitochondrial biogenesis and dynamics. We show that hippocampal pyramidal neurons in mutUNG1 mice, as well as cultured rat hippocampal neurons and human fibroblasts with H2O2 induced oxidative stress, improve markers of mitochondrial biogenesis, dynamics and function when fed on a KD, and when exposed to the ketone body β-hydroxybutyrate, respectively, by upregulating PGC1α, SIRT3 and UCP2, and (in cultured cells) increasing the oxygen consumption rate (OCR) and the NAD+/NADH ratio. The mitochondrial level of UCP2 was significantly higher in the perikarya and axon terminals of hippocampus CA1 pyramidal neurons in KD treated mutUNG1 mice compared with mutUNG1 mice fed a standard diet. The β-hydroxybutyrate receptor GPR109a (HCAR2), but not the structurally closely related lactate receptor GPR81 (HCAR1), was upregulated in mutUNG1 mice on a KD, suggesting a selective influence of KD on ketone body receptor mechanisms. We conclude that progressive mitochondrial dysfunction in mutUNG1 expressing mice causes oxidative stress, and that exposure of animals to KD, or of cells to ketone body in vitro, elicits compensatory mechanisms acting to augment mitochondrial mass and bioenergetics via the PGC1α-SIRT3-UCP2 axis (The compensatory processes are overwhelmed in the mutUNG1 mice by all the newly formed mitochondria being dysfunctional).
Collapse
Affiliation(s)
- Md Mahdi Hasan-Olive
- Synaptic Neurochemistry and Amino Acid Transporter Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway. .,Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway. .,Center for Healthy Aging, Department of Neurosciences and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Knut H Lauritzen
- Synaptic Neurochemistry and Amino Acid Transporter Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Mohammad Ali
- Department of Biochemistry, Sir Salimullah Medical College & Mitford Hospital, Dhaka, Bangladesh
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Neurosciences and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jon Storm-Mathisen
- Synaptic Neurochemistry and Amino Acid Transporter Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Linda H Bergersen
- Synaptic Neurochemistry and Amino Acid Transporter Laboratory, Division of Anatomy and CMBN/SERTA Healthy Brain Ageing Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway. .,Brain and Muscle Energy Group, Electron Microscopy Laboratory, Institute of Oral Biology, University of Oslo, Oslo, Norway. .,Center for Healthy Aging, Department of Neurosciences and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
33
|
Zheng Y, Liu T, Wang Z, Xu Y, Zhang Q, Luo D. Low molecular weight fucoidan attenuates liver injury via SIRT1/AMPK/PGC1α axis in db/db mice. Int J Biol Macromol 2018; 112:929-936. [DOI: 10.1016/j.ijbiomac.2018.02.072] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/31/2018] [Accepted: 02/11/2018] [Indexed: 02/06/2023]
|
34
|
Mozos I, Stoian D, Caraba A, Malainer C, Horbańczuk JO, Atanasov AG. Lycopene and Vascular Health. Front Pharmacol 2018; 9:521. [PMID: 29875663 PMCID: PMC5974099 DOI: 10.3389/fphar.2018.00521] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 04/30/2018] [Indexed: 01/20/2023] Open
Abstract
Lycopene is a lipophilic, unsaturated carotenoid, found in red-colored fruits and vegetables, including tomatoes, watermelon, papaya, red grapefruits, and guava. The present work provides an up to date overview of mechanisms linking lycopene in the human diet and vascular changes, considering epidemiological data, clinical studies, and experimental data. Lycopene may improve vascular function and contributes to the primary and secondary prevention of cardiovascular disorders. The main activity profile of lycopene includes antiatherosclerotic, antioxidant, anti-inflammatory, antihypertensive, antiplatelet, anti-apoptotic, and protective endothelial effects, the ability to improve the metabolic profile, and reduce arterial stiffness. In this context, lycopene has been shown in numerous studies to exert a favorable effect in patients with subclinical atherosclerosis, metabolic syndrome, hypertension, peripheral vascular disease, stroke and several other cardiovascular disorders, although the obtained results are sometimes inconsistent, which warrants further studies focusing on its bioactivity.
Collapse
Affiliation(s)
- Ioana Mozos
- Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, Timiṣoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babes” University of Medicine and Pharmacy, Timiṣoara, Romania
| | - Dana Stoian
- 2nd Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, Timiṣoara, Romania
| | - Alexandru Caraba
- 1st Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, Timiṣoara, Romania
| | | | - Jarosław O. Horbańczuk
- Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Integrative analysis of indirect calorimetry and metabolomics profiling reveals alterations in energy metabolism between fed and fasted pigs. J Anim Sci Biotechnol 2018; 9:41. [PMID: 29796254 PMCID: PMC5956531 DOI: 10.1186/s40104-018-0257-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/19/2018] [Indexed: 11/16/2022] Open
Abstract
Background Fasting is a simple metabolic strategy that is used to estimate the maintenance energy requirement where the energy supply for basic physiological functions is provided by the mobilization of body reserves. However, the underlying metabolic components of maintenance energy expenditure are not clear. This study investigated the differences in heat production (HP), respiratory quotient (RQ) and plasma metabolites in pigs in the fed and fasted state, using the techniques of indirect calorimetry and metabolomics. Methods Nine barrows (45.2 ± 1.7 kg BW) were fed corn-soybean based meal diets and were kept in metabolism crates for a period of 14 d. After 7 d adaptation, pigs were transferred to respiratory chambers to determine HP and RQ based on indirect calorimetry. Pigs were fed the diet at 2,400 kJ ME/(kg BW0.6·d) during d 8 to 12. The last 2 d were divided into 24 h fasting and 48 h fasting treatment, respectively. Plasma samples of each pig were collected from the anterior vena cava during the last 3 d (1 d while pigs were fed and 2 d during which they were fasted). The metabolites of plasma were determined by high-resolution mass spectrometry using a metabolomics approach. Results Indirect calorimetry analysis revealed that HP and RQ were no significant difference between 24 h fasting and 48 h fasting, which were lower than those of fed state (P < 0.01). The nitrogen concentration of urine tended to decrease with fasting (P = 0.054). Metabolomics analysis between the fed and fasted state revealed differences in 15 compounds, most of which were not significantly different between 24 h fasting and 48 h fasting. Identified compounds were enriched in metabolic pathways related to linoleic acid metabolism, amino acid metabolism, sphingolipid metabolism, and pantothenate and CoA biosynthesis. Conclusion These results suggest that the decreases in HP and RQ of growing pigs under fasting conditions were associated with the alterations of linoleic acid metabolism and amino acid metabolism. The integrative analysis also revealed that growing pigs under a 24-h fasting were more appropriate than a 48-h fasting to investigate the metabolic components of maintenance energy expenditure.
Collapse
|
36
|
Shen P, Kershaw JC, Yue Y, Wang O, Kim KH, McClements DJ, Park Y. Effects of conjugated linoleic acid (CLA) on fat accumulation, activity, and proteomics analysis in Caenorhabditis elegans. Food Chem 2018; 249:193-201. [PMID: 29407924 DOI: 10.1016/j.foodchem.2018.01.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 12/27/2017] [Accepted: 01/02/2018] [Indexed: 12/14/2022]
Abstract
Conjugated linoleic acid (CLA) has been reported to reduce fat storage in cell culture and animal models. In the current study, the effects of CLA on the fat accumulation, activities, and proteomics were investigated using Caenorhabditis elegans. 100 µM CLA-TG nanoemulsion significantly reduced fat accumulation by 29% compared to linoleic acid (LA)-TG treatment via sir-2.1 (the ortholog of Sirtuin 1), without altering the worm size, growth rate, and pumping rate of C. elegans. CLA significantly increased moving speed and amplitude (the average centroid displacement over the entire track) of wild type worms compared to the LA group and these effects were dependent on aak-2 (AMPKα ortholog) and sir-2.1. Proteomics analysis showed CLA treatment influences various proteins associated in reproduction and development, translation, metabolic processes, and catabolism and proteolysis, in C. elegans. We have also confirmed the proteomics data that CLA reduced the fat accumulation via abs-1 (ATP Synthase B homolog). However, there were no significant effects of CLA on brood size, progeny numbers, and hatchability compared to LA treatment.
Collapse
Affiliation(s)
- Peiyi Shen
- Department of Food Science, University of Massachusetts, Amherst, USA
| | - Jonathan C Kershaw
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Yiren Yue
- Department of Food Science, University of Massachusetts, Amherst, USA
| | - Ou Wang
- Department of Food Science, University of Massachusetts, Amherst, USA; National Institute for Nutrition and Health, Chinese Centre for Disease Control and Prevention, Beijing 10050, China
| | - Kee-Hong Kim
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA
| | | | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, USA.
| |
Collapse
|
37
|
Supplementation with an insoluble fiber obtained from carob pod (Ceratonia siliqua L.) rich in polyphenols prevents dyslipidemia in rabbits through SIRT1/PGC-1α pathway. Eur J Nutr 2017; 58:357-366. [DOI: 10.1007/s00394-017-1599-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023]
|
38
|
Chen GC, Chen WH, Tseng KT, Chao PM. The anti-adiposity effect of bitter melon seed oil is solely attributed to its fatty acid components. Lipids Health Dis 2017; 16:186. [PMID: 28962621 PMCID: PMC5622538 DOI: 10.1186/s12944-017-0578-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/22/2017] [Indexed: 11/10/2022] Open
Abstract
Background Obesity is the leading chronic disease affecting people of all ages. The objective of this study was to optimize composition of a bitter melon seed oil (BMSO) product to maximize its anti-adiposity effect. Methods Bleaching oil, saponifiables and non-saponifiables were prepared from BMSO, with α-eleostearic acid (α-ESA) content in BMSO maintained in bleaching oil and saponifiables. C57BL/6 J mice were allocated into five groups (n = 10/group) to receive diet C [30% soybean oil (SBO)], BM [25% SBO + 5% BMSO], BMS, BMNS or BMD. For the three latter diets, saponifiables (hydrolyzed fatty acids from BMSO), non-saponifiables (excluding fatty acids from BMSO) or bleaching oil (excluding pigments from BMSO), respectively, were added in amount equivalent to their content in 5% BMSO and SBO was added to bring total fat to 30%. After 14 wk., indices associated with adiposity and safety, as well as lipid metabolic signaling in white adipose tissue (WAT), were measured. Results The body fat percentage of mice in group BM, BMS, BMNS, and BMD were 90 ± 26, 76 ± 21, 115 ± 30 and 95 ± 17% of that in group C. Based on body fat percentage and plasma leptin concentrations, an anti-adiposity effect was evident in groups BM, BMS and BMD (greatest effect in BMS). Histologically, inguinal fat had smaller adipocytes in groups BM, BMS and BMD (P < 0.05), but not in group BMNS, relative to group C. There were no differences among groups in blood pressure or heart rate. Moreover, Sirt1 mRNA levels in inguinal fat were significantly greater in groups BM, BMS and BMD than group C. Conclusion We concluded that the anti-adiposity function of BMSO was solely attributed to the fatty acid fraction, with the free fatty acid form having the greatest effect. Electronic supplementary material The online version of this article (10.1186/s12944-017-0578-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gou-Chun Chen
- Department of Nutrition, China Medical University, Taichung, 404, Taiwan
| | | | | | - Pei-Min Chao
- Department of Nutrition, China Medical University, Taichung, 404, Taiwan.
| |
Collapse
|
39
|
Altered gene expression of epigenetic modifying enzymes in response to dietary supplementation with linseed oil. J DAIRY RES 2017; 84:119-123. [PMID: 28524020 DOI: 10.1017/s002202991700022x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Recently we showed that 5% linseed oil (LSO) and 5% safflower oil (SFO) supplementation of cow's diets reduced milk fat yield by 30·38 and 32·42% respectively, accompanied by differential expression of genes and regulation by microRNAs (miRNA). This research communication addresses the hypothesis that epigenetic regulation could be involved in the observed milk fat reduction. Thus, this study investigated the gene expression pattern of major epigenetic modifying enzymes in response to dietary supplementation with LSO or SFO. Twenty-six Canadian Holstein cows in mid lactation were randomly assigned to two groups (13/group) and fed a control diet for 28 d (day -28 to -1) (control period- CP) followed by a treatment period (TP) (control diet supplemented with 5% LSO (LSO treatment) or 5% SFO (SFO treatment) of 28 d (day +1 to +28). After treatment, cows in the two groups were returned to the control diet for another 28 d (day +29 to +56) (post treatment period-PTP). Milk samples were collected on day -1 (CP), +7, +28 (TP) and +56 (PTP) for RNA isolation and measurement of the expression of thirteen epigenetic modifying genes including two DNA methytrasferases (DNMT1, DNMT3A), four histone acetylases (HAT1, KAT2A, KAT5 and CREBBP), five histone deacetylases (HDAC1, HDAC2, HDAC3, SIRT1 and SIRT2) and two histone methytransferases (EHMT2 and PRMT1) by qPCR. Linseed oil supplementation significantly repressed the expression of EHMT2, HDAC2 and HDAC3 on day +7 (P < 0·05) and KAT2A and SIRT2 on day +28 (P < 0·05) as compared with the control period (day -1) while SFO had no effect. When LSO was withdrawn, the expression of some of the genes increased slightly but did not reach control (day -1) levels at the end of the PTP. Our study demonstrated a significant role of LSO in the epigenetic regulation of fatty acid synthesis as compared to SFO. The effect of LSO may be related to its higher degree of unsaturation and might represent a different regulatory mechanism which needs further investigation.
Collapse
|
40
|
Pande S, Kratasyuk VA, Medvedeva NN, Kolenchukova OA, Salmina AB. Nutritional biomarkers: Current view and future perspectives. Crit Rev Food Sci Nutr 2017; 58:3055-3069. [PMID: 28678523 DOI: 10.1080/10408398.2017.1350136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a poor relationship between nutrient intake and existing nutritional biomarkers due to variety of factors affecting their sensitivity and specificity. To explore the impact of nutrients at molecular level and devising a sensitive biomarker, proteomics is a central technology with sirtuins as one of the most promising nutritional biomarker. Sirtuins (seven mammalian sirtuins reported so far) have been reported to perform protein deacetylases and ADP-ribosyltransferases activity. It is distributed in different cellular compartments thereby controlling several metabolic processes. Sirtuins are oxidized nicotinamide adenine dinucleotide dependent, which implicates a direct effect of the metabolic state of the cell on its activity. Calorie restriction upregulates the mammalian sirtuin protein levels in variety of tissues and organs where it acts upon both histone and nonhistone substrates. Sirtuin senses nutrient availability and impacts gluconeogenesis, glycolysis, and insulin sensitivity. It deacetylates and inhibits the nuclear receptor that activates fat synthesis and adipogenesis in the body, leading to fat loss and bringing favorable cellular and health changes. Sirtuins mediates intracellular response that promotes cell survival, DNA damage repair thereby increasing the cell longitivity. The activation of sirtuins brings a wide spectrum of other health benefits and its activity levels are indicative of nutritional status as well as disease progression in cancer, inflammation, obesity, cardiovascular diseases, and viral infections. There are several foods that activate sirtuin activity and offer significant health benefits by their consumption.
Collapse
Affiliation(s)
- Shubhra Pande
- a Laboratory of Bioluminescent Biotechnologies, Department of Biophysics , Institute of Fundamental Biology and Biotechnology, Siberian Federal University , Krasnoyarsk , Russia.,b Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky , Krasnoyarsk , Russia
| | - Valentina A Kratasyuk
- a Laboratory of Bioluminescent Biotechnologies, Department of Biophysics , Institute of Fundamental Biology and Biotechnology, Siberian Federal University , Krasnoyarsk , Russia.,c Federal State Budgetary Scientific Institution "Institute of Biophysics, Siberian Branch of RAS" , Krasnoyarsk , Russia
| | - Nadezhda N Medvedeva
- b Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky , Krasnoyarsk , Russia
| | - Oxana A Kolenchukova
- a Laboratory of Bioluminescent Biotechnologies, Department of Biophysics , Institute of Fundamental Biology and Biotechnology, Siberian Federal University , Krasnoyarsk , Russia.,d Federal State Budgetary Scientific Institution "Scientific Research Institute of medical problems of the North" , Krasnoyarsk , Russia
| | - Alla B Salmina
- b Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky , Krasnoyarsk , Russia
| |
Collapse
|
41
|
de Melo LGP, Nunes SOV, Anderson G, Vargas HO, Barbosa DS, Galecki P, Carvalho AF, Maes M. Shared metabolic and immune-inflammatory, oxidative and nitrosative stress pathways in the metabolic syndrome and mood disorders. Prog Neuropsychopharmacol Biol Psychiatry 2017; 78:34-50. [PMID: 28438472 DOI: 10.1016/j.pnpbp.2017.04.027] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/27/2017] [Accepted: 04/08/2017] [Indexed: 02/08/2023]
Abstract
This review examines the shared immune-inflammatory, oxidative and nitrosative stress (IO&NS) and metabolic pathways underpinning metabolic syndrome (MetS), bipolar disorder (BD) and major depressive disorder (MDD). Shared pathways in both MetS and mood disorders are low grade inflammation, including increased levels of pro-inflammatory cytokines and acute phase proteins, increased lipid peroxidation with formation of malondialdehyde and oxidized low density lipoprotein cholesterol (LDL-c), hypernitrosylation, lowered levels of antioxidants, most importantly zinc and paraoxonase (PON1), increased bacterial translocation (leaky gut), increased atherogenic index of plasma and Castelli risk indices; and reduced levels of high-density lipoprotein (HDL-c) cholesterol. Insulin resistance is probably not a major factor associated with mood disorders. Given the high levels of IO&NS and metabolic dysregulation in BD and MDD and the high comorbidity with the atherogenic components of the MetS, mood disorders should be viewed as systemic neuro-IO&NS-metabolic disorders. The IO&NS-metabolic biomarkers may have prognostic value and may contribute to the development of novel treatments targeting neuro-immune, neuro-oxidative and neuro-nitrosative pathways.
Collapse
Affiliation(s)
- Luiz Gustavo Piccoli de Melo
- Department of Clinical Medicine, Londrina State University (UEL), Health Sciences Centre, Londrina, Paraná, Brazil; Center of Approach and Treatment for Smokers, University Hospital, Londrina State University, University Campus, Londrina, Paraná, Brazil; Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Sandra Odebrecht Vargas Nunes
- Department of Clinical Medicine, Londrina State University (UEL), Health Sciences Centre, Londrina, Paraná, Brazil; Center of Approach and Treatment for Smokers, University Hospital, Londrina State University, University Campus, Londrina, Paraná, Brazil; Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | | | - Heber Odebrecht Vargas
- Department of Clinical Medicine, Londrina State University (UEL), Health Sciences Centre, Londrina, Paraná, Brazil; Center of Approach and Treatment for Smokers, University Hospital, Londrina State University, University Campus, Londrina, Paraná, Brazil; Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Décio Sabbattini Barbosa
- Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil; Department of Clinical and Toxicological Analysis, State University of Londrina, Londrina, Paraná, Brazil
| | - Piotr Galecki
- Department of Adult Psychiatry, University of Lodz, Lodz, Poland
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- Health Sciences Graduation Program, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil; Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Plovdiv University, Plovdiv, Bulgaria; Revitalis, Waalre, The Netherlands; Impact Strategic Research Center, Deakin University, Geelong, Australia.
| |
Collapse
|
42
|
Ding RB, Bao J, Deng CX. Emerging roles of SIRT1 in fatty liver diseases. Int J Biol Sci 2017; 13:852-867. [PMID: 28808418 PMCID: PMC5555103 DOI: 10.7150/ijbs.19370] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/19/2017] [Indexed: 12/11/2022] Open
Abstract
Fatty liver diseases, which are commonly associated with high-fat/calorie diet, heavy alcohol consumption and/or other metabolic disorder causes, lead to serious medical concerns worldwide in recent years. It has been demonstrated that metabolic homeostasis disruption is most likely to be responsible for this global epidemic. Sirtuins are a group of conserved nicotinamide adenine dinucleotide (NAD+) dependent histone and/or protein deacetylases belonging to the silent information regulator 2 (Sir2) family. Among seven mammalian sirtuins, sirtuin 1 (SIRT 1) is the most extensively studied one and is involved in both alcoholic and nonalcoholic fatty liver diseases. SIRT1 plays beneficial roles in regulating hepatic lipid metabolism, controlling hepatic oxidative stress and mediating hepatic inflammation through deacetylating some transcriptional regulators against the progression of fatty liver diseases. Here we summarize the latest advances of the biological roles of SIRT1 in regulating lipid metabolism, oxidative stress and inflammation in the liver, and discuss the potential of SIRT1 as a therapeutic target for treating alcoholic and nonalcoholic fatty liver diseases.
Collapse
Affiliation(s)
- Ren-Bo Ding
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Jiaolin Bao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Chu-Xia Deng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| |
Collapse
|
43
|
Bolívar BE, Welch JT. Studies of the Binding of Modest Modulators of the Human Enzyme, Sirtuin 6, by STD NMR. Chembiochem 2017; 18:931-940. [PMID: 28222243 DOI: 10.1002/cbic.201600655] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Indexed: 01/02/2023]
Abstract
Pyrazinamide (PZA), an essential constituent of short-course tuberculosis chemotherapy, binds weakly but selectively to Sirtuin 6 (SIRT6). Despite the structural similarities between nicotinamide (NAM), PZA, and pyrazinoic acid (POA), these inhibitors modulate SIRT6 by different mechanisms and through different binding sites, as suggested by saturation transfer difference (STD) NMR. Available experimental evidence, such as that derived from crystal structures and kinetic experiments, has been of only limited utility in elucidation of the mechanistic details of sirtuin inhibition by NAM or other inhibitors. For instance, crystallographic structural analysis of sirtuin binding sites does not help us understand important differences in binding affinities among sirtuins or capture details of such dynamic process. Hence, STD NMR was utilized throughout this study. Our results not only agreed with the binding kinetics experiments but also gave a qualitative insight into the binding process. The data presented herein suggested some details about the geometry of the binding epitopes of the ligands in solution with the apo- and holoenzyme. Recognition that SIRT6 is affected selectively by PZA, an established clinical agent, suggests that the rational development of more potent and selective NAM surrogates might be possible. These derivatives might be accessible by employing the malleability of this scaffold to assist in the identification by STD NMR of the motifs that interact with the apo- and holoenzymes in solution.
Collapse
Affiliation(s)
- Beatriz E Bolívar
- Department of Chemistry, University at Albany, 1400 Washington Avenue, Albany, NY, 12205, USA
| | - John T Welch
- Department of Chemistry, University at Albany, 1400 Washington Avenue, Albany, NY, 12205, USA
| |
Collapse
|
44
|
Lee AMC, Shandala T, Soo PP, Su YW, King TJ, Chen KM, Howe PR, Xian CJ. Effects of Resveratrol Supplementation on Methotrexate Chemotherapy-Induced Bone Loss. Nutrients 2017; 9:nu9030255. [PMID: 28282956 PMCID: PMC5372918 DOI: 10.3390/nu9030255] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/08/2017] [Indexed: 12/27/2022] Open
Abstract
Intensive cancer chemotherapy is known to cause bone defects, which currently lack treatments. This study investigated the effects of polyphenol resveratrol (RES) in preventing bone defects in rats caused by methotrexate (MTX), a commonly used antimetabolite in childhood oncology. Young rats received five daily MTX injections at 0.75 mg/kg/day. RES was orally gavaged daily for seven days prior to, and during, five-day MTX administration. MTX reduced growth plate thickness, primary spongiosa height, trabecular bone volume, increased marrow adipocyte density, and increased mRNA expression of the osteogenic, adipogenic, and osteoclastogenic factors in the tibial bone. RES at 10 mg/kg was found not to affect bone health in normal rats, but to aggravate the bone damage in MTX-treated rats. However, RES supplementation at 1 mg/kg preserved the growth plate, primary spongiosa, bone volume, and lowered the adipocyte density. It maintained expression of genes involved in osteogenesis and decreased expression of adipogenic and osteoclastogenic factors. RES suppressed osteoclast formation ex vivo of bone marrow cells from the treated rats. These data suggest that MTX can enhance osteoclast and adipocyte formation and cause bone loss, and that RES supplementation at 1 mg/kg may potentially prevent these bone defects.
Collapse
Affiliation(s)
- Alice M C Lee
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Tetyana Shandala
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Pei Pei Soo
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Yu-Wen Su
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Tristan J King
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Ke-Ming Chen
- Institute of Orthopaedics, Lanzhou General Hospital, Lanzhou Command of People's Liberation Army, Lanzhou 730050, China.
| | - Peter R Howe
- Clinical Nutrition Research Centre, University of Newcastle, Callaghan NSW 2308, Australia.
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia.
| |
Collapse
|
45
|
Lee M, Choi S, Lee Y, Oh HH. The Gender Association of the SIRT1 rs7895833 Polymorphism with Pediatric Obesity: A 3-Year Panel Study. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2017; 9:265-275. [PMID: 28118644 DOI: 10.1159/000454713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/24/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Sirtuin 1 (SIRT1), a longevity-associated gene, has pleiotropic functions. We investigated whether SIRT1 variation is associated with pediatric obesity. METHODS During 3 years of follow-up of 219 children (101 boys, 118 girls) aged 8 or 9 years at baseline, obesity parameters such as anthropometrics, plasma lipid and insulin resistance profiles, and nutrient intakes were analyzed with regard to 3 genotypes of SIRT1 rs7895833 (GG, GA, and AA). RESULTS The prevalence of obesity including overweight had increased from 18.3% (in 2007) to 25.1% (in 2010), and the incidence of obesity over 3 years from nonobesity at the baseline was 11.7%. In the obesity group (BMI >85th percentile) that had been nonobese 3 years before, the frequency of the GA+AA genotypes was higher than that of the GG genotype. Among the total number of subjects, the values for criteria for obesity such as BMI and waist circumference were higher in the GA+AA group than in the GG group. In boys, the reductions in total cholesterol and low-density lipoprotein cholesterol levels in the GG group were considerably greater than those in the GA+AA group, even though the changes in carbohydrate, fat, and protein intake in the GG group were higher than in the GA+AA group. In girls, the reductions in fasting blood sugar and homeostatic model assessment insulin resistance (HOMA-IR) levels were greater in the GA+AA group than in the GG group, despite unchanged energy intakes over 3 years. CONCLUSIONS We identified an association between SIRT1 variation and pediatric obesity in Korean children with a gender difference.
Collapse
Affiliation(s)
- Myoungsook Lee
- Department of Food and Nutrition, Sungshin Women's University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
46
|
Glucagon-like peptide-1 effects lipotoxic oxidative stress by regulating the expression of microRNAs. Biochem Biophys Res Commun 2016; 482:1462-1468. [PMID: 27956176 DOI: 10.1016/j.bbrc.2016.12.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 12/08/2016] [Indexed: 12/24/2022]
Abstract
Aim to confirm whether the treatment of GLP-1 can modulated body weight, lipid metabolism, insulin content, pancreas oxidative stress, improved T-AOC, MDA levels related to FFA-Induced oxidative stress in C57BL/6 mice and INS-1 cells. In this study, GLP-1 makes the expression of AMPK, PPARα, CPT1A and SIRT1 increased, and the expression of SREBP1c, miR-33 and miR-370 decreased. Interestingly, the effects of GLP-1 were less dose dependent as GLP-1 regulated the FFA, which related to gene expression at much lower doses (3 μg/kg, 10 mM, mice and INS-1 respectively) and effects were relatively maintained at higher dose (30 μg/kg, 100 mM, mice and INS-1 respectively) as well. Subsequently, the analysis showed that inhibited expression of miR-33 and miR-370 upregulated the expression of CPT1A and SIRT1, reversely mimics. These results demonstrated for the first time that GLP-1 improve lipotoxic oxidative stress of pancreas by regulate expression of microRNAs.
Collapse
|
47
|
Ren J, Xu N, Ma Z, Li Y, Li C, Wang Y, Tian Y, Liu X, Kang X. Characteristics of expression and regulation of sirtuins in chicken (Gallus gallus). Genome 2016; 60:431-440. [PMID: 28177837 DOI: 10.1139/gen-2016-0125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sirtuins (SIRT1-SIRT7) are a family of NAD+-dependent protein deacetylases that are linked to post-translational regulation of many metabolic processes. There are few reports available for chicken sirtuins (designated cSIRT1-cSIRT7), whose expression and regulation in the liver have yet to be explored. In the present study, we characterized the expression and regulation of sirtuin family members in chicken liver. The results showed that the sirtuin family members in chicken share the same conserved functional SIR2 domains. All the sirtuin family members were expressed extensively in all tissues examined, and the expression levels of cSIRT1, cSIRT2, cSIRT4, cSIRT6, and cSIRT7 in the liver increased significantly with sexual maturity. However, all sirtuin family members were downregulated (P < 0.05) in chicken livers and cultured primary hepatocytes treated with 17β-estradiol. We concluded that the expression levels of some chicken sirtuin family members in the liver were upregulated with sexual maturation, but might not be regulated directly by estrogen. Whereas estrogen could be used as an inhibitor of all sirtuins, both in vivo and in vitro.
Collapse
Affiliation(s)
- Junxiao Ren
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Naiyi Xu
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Zheng Ma
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Yanmin Li
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Cuicui Li
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China.,b Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450002, China.,c International Joint Research Laboratory for Poultry Breeding of Henan, Henan, Zhengzhou 450002, China
| | - Yanbin Wang
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China.,b Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450002, China.,c International Joint Research Laboratory for Poultry Breeding of Henan, Henan, Zhengzhou 450002, China
| | - Yadong Tian
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China.,b Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450002, China.,c International Joint Research Laboratory for Poultry Breeding of Henan, Henan, Zhengzhou 450002, China
| | - Xiaojun Liu
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China.,b Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450002, China.,c International Joint Research Laboratory for Poultry Breeding of Henan, Henan, Zhengzhou 450002, China
| | - Xiangtao Kang
- a College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China.,b Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou 450002, China.,c International Joint Research Laboratory for Poultry Breeding of Henan, Henan, Zhengzhou 450002, China
| |
Collapse
|
48
|
AAV-mediated Sirt1 overexpression in skeletal muscle activates oxidative capacity but does not prevent insulin resistance. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 5:16072. [PMID: 27909699 PMCID: PMC5111573 DOI: 10.1038/mtm.2016.72] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/15/2016] [Accepted: 09/29/2016] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes is characterized by triglyceride accumulation and reduced lipid oxidation capacity in skeletal muscle. SIRT1 is a key protein in the regulation of lipid oxidation and its expression is reduced in the skeletal muscle of insulin resistant mice. In this tissue, Sirt1 up-regulates the expression of genes involved in oxidative metabolism and improves mitochondrial function mainly through PPARGC1 deacetylation. Here we examined whether Sirt1 overexpression mediated by adeno-associated viral vectors of serotype 1 (AAV1) specifically in skeletal muscle can counteract the development of insulin resistance induced by a high fat diet in mice. AAV1-Sirt1-treated mice showed up-regulated expression of key genes related to β-oxidation together with increased levels of phosphorylated AMP protein kinase. Moreover, SIRT1 overexpression in skeletal muscle also increased basal phosphorylated levels of AKT. However, AAV1-Sirt1 treatment was not enough to prevent high fat diet-induced obesity and insulin resistance. Although Sirt1 gene transfer to skeletal muscle induced changes at the muscular level related with lipid and glucose homeostasis, our data indicate that overexpression of SIRT1 in skeletal muscle is not enough to improve whole-body insulin resistance and that suggests that SIRT1 has to be increased in other metabolic tissues to prevent insulin resistance.
Collapse
|
49
|
Kurylowicz A. In Search of New Therapeutic Targets in Obesity Treatment: Sirtuins. Int J Mol Sci 2016; 17:ijms17040572. [PMID: 27104517 PMCID: PMC4849028 DOI: 10.3390/ijms17040572] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 03/29/2016] [Accepted: 04/06/2016] [Indexed: 12/11/2022] Open
Abstract
Most of the available non-invasive medical therapies for obesity are non-efficient in a long-term evaluation; therefore there is a constant need for new methods of treatment. Research on calorie restriction has led to the discovery of sirtuins (silent information regulators, SIRTs), enzymes regulating different cellular pathways that may constitute potential targets in the treatment of obesity. This review paper presents the role of SIRTs in the regulation of glucose and lipid metabolism as well as in the differentiation of adipocytes. How disturbances of SIRTs’ expression and activity may lead to the development of obesity and related complications is discussed. A special emphasis is placed on polymorphisms in genes encoding SIRTs and their possible association with susceptibility to obesity and metabolic complications, as well as on data regarding altered expression of SIRTs in human obesity. Finally, the therapeutic potential of SIRTs-targeted strategies in the treatment of obesity and related disorders is discussed.
Collapse
Affiliation(s)
- Alina Kurylowicz
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland.
| |
Collapse
|
50
|
Caglayan EK, Engin-Ustun Y, Gocmen AY, Sarı N, Seckin L, Kara M, Polat MF. Is there any relationship between serum sirtuin-1 level and neutrophil-lymphocyte ratio in hyperemesis gravidarum? J Perinat Med 2016; 44:315-20. [PMID: 26352076 DOI: 10.1515/jpm-2015-0178] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/29/2015] [Indexed: 12/20/2022]
Abstract
AIM The aim of this study was to evaluate the relationship between serum sirtuin-1 (SIRT1) level and neutrophil-lymphocyte ratio (NLR) with hyperemesis gravidarum (HG). METHODS Overall, 90 patients who presented with pregnancy between August 2013 and November 2014 were included in the study. The patients were divided into two groups: patients with HG (n=45) and patients without HG (control group [C]; n=45). The patients with comorbid conditions other than pregnancy (disease or medication) were excluded. In all patients, demographic data including age, body mass index (BMI), gestational week, and smoking status were recorded. Blood samples were drawn for complete blood count and measurements of blood lipid, liver enzymes, serum SIRT1, and insulin levels. NLR was calculated from CBC. RESULTS No significant differences were detected in age, BMI, or GA between groups (P>0.05). Serum SIRT1 and NLR were found to be significantly higher in patients with HG compared with those in the control group (P=0.001 and 0.006, respectively). CONCLUSION In HG, both SIRT1 level and NLR increased. In HG, this occurred as a response to metabolic alterations and potential inflammation.
Collapse
|