51
|
Zhang Q, Ding Y, Zhang J, Wang L. EFFECTS OF ACUTE EXERCISE WITH DIFFERENT INTENSITIES ON GLYCEMIC CONTROL IN PATIENTS WITH TYPE 2 DIABETES MELLITUS. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2021; 17:212-218. [PMID: 34925570 PMCID: PMC8665237 DOI: 10.4183/aeb.2021.212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Exercise intensity is one of the most important factors that determines the effects of exercise; however, there is little known about the acute glycemic control of different exercise intensities on patients with Type 2 Diabetes Mellitus (T2DM). Here we aimed at exploring the influence of a single bout of exercise with different intensities on blood glucose levels in T2DM patients. METHODS Fifteen subjects (54.7 ± 5.8 years old) participated in a session of walking (WG), jogging (JG), or sedentary control (CG) in a randomized order on three different days. Distances in both WG and JG were set as 2 Km with a speed set as 4~4.5 Km/h for walking and 5~6 Km/h for jogging based on pretrial test. Blood glucose levels at fasting (~6:30am), pre-exercise (~8:30am), post-exercise (~9am), 11am and 4pm were detected. RESULTS Walking and jogging reached approximately moderate and high intensity based on the immediate post-exercise heart rate and RPE scores. Blood glucose levels at fasting, pre-exercise and 4pm were not substantially different among all groups (p > 0.05). JG had a significantly lower post-exercise blood glucose level (p < 0.05) when compared with CG and WG. The blood glucose level at 11am was notably lower in WG and JG than in CG (p < 0.05). CONCLUSIONS Both a single bout of jogging and walking can lower postprandial blood glucose levels in T2DM patients. When matched for exercise distance, jogging represents a more effective strategy to immediately lower postprandial glucose levels than walking.
Collapse
Affiliation(s)
- Q.Q. Zhang
- First Affiliated Hospital of Anhui Medical University - Department of Pediatrics, Hefei, Anhui, China
- Soochow University - Medical College, School of Nursing, Suzhou, Jiangsu, China
| | - Y.J. Ding
- Soochow University - Medical College, School of Nursing, Suzhou, Jiangsu, China
- Changshu No.2 People’s Hospital - Department of Endocrinology, Changshu, Jiangsu, China
| | - J.J. Zhang
- Soochow University - Medical College, School of Nursing, Suzhou, Jiangsu, China
- Zhejiang University School of Medicine Sir Run Run Shaw Hospital - Intensive Care Unit, Hangzhou, Zhejiang China
| | - L. Wang
- Soochow University - Medical College, School of Nursing, Suzhou, Jiangsu, China
| |
Collapse
|
52
|
Queiroz AL, Lessard SJ, Ouchida AT, Araujo HN, Gonçalves DA, Simões Fróes Guimarães DSP, Teodoro BG, So K, Espreafico EM, Hirshman MF, Alberici LC, Kettelhut IDC, Goodyear LJ, Silveira LR. The MicroRNA miR-696 is regulated by SNARK and reduces mitochondrial activity in mouse skeletal muscle through Pgc1α inhibition. Mol Metab 2021; 51:101226. [PMID: 33812060 PMCID: PMC8121711 DOI: 10.1016/j.molmet.2021.101226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE MicroRNAs (miRNA) are known to regulate the expression of genes involved in several physiological processes including metabolism, mitochondrial biogenesis, proliferation, differentiation, and cell death. METHODS Using "in silico" analyses, we identified 219 unique miRNAs that potentially bind to the 3'UTR region of a critical mitochondrial regulator, the peroxisome proliferator-activated receptor gamma coactivator (PGC) 1 alpha (Pgc1α). Of the 219 candidate miRNAs, miR-696 had one of the highest interactions at the 3'UTR of Pgc1α, suggesting that miR-696 may be involved in the regulation of Pgc1α. RESULTS Consistent with this hypothesis, we found that miR-696 was highly expressed in the skeletal muscle of STZ-induced diabetic mice and chronic high-fat-fed mice. C2C12 muscle cells exposed to palmitic acid also exhibited a higher expression of miR-696. This increased expression corresponded with a reduced expression of oxidative metabolism genes and reduced mitochondrial respiration. Importantly, reducing miR-696 reversed decreases in mitochondrial activity in response to palmitic acid. Using C2C12 cells treated with the AMP-activated protein kinase (AMPK) activator AICAR and skeletal muscle from AMPKα2 dominant-negative (DN) mice, we found that the signaling mechanism regulating miR-696 did not involve AMPK. In contrast, overexpression of SNF1-AMPK-related kinase (SNARK) in C2C12 cells increased miR-696 transcription while knockdown of SNARK significantly decreased miR-696. Moreover, muscle-specific transgenic mice overexpressing SNARK exhibited a lower expression of Pgc1α, elevated levels of miR-696, and reduced amounts of spontaneous activity. CONCLUSIONS Our findings demonstrate that metabolic stress increases miR-696 expression in skeletal muscle cells, which in turn inhibits Pgc1α, reducing mitochondrial function. SNARK plays a role in this process as a metabolic stress signaling molecule inducing the expression of miR-696.
Collapse
Affiliation(s)
- André L Queiroz
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil; Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah J Lessard
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Amanda T Ouchida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Hygor N Araujo
- Obesity and Comorbidities Research Center, OCRC, IB, UNICAMP, Campinas, Brazil
| | - Dawit A Gonçalves
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | | | - Bruno G Teodoro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil; Department of Physics and Chemistry, Faculty of Pharmaceutical Science, USP, Ribeirão Preto, Brazil
| | - Kawai So
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Enilza M Espreafico
- Department of Cell Biology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Michael F Hirshman
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Luciane C Alberici
- Department of Physics and Chemistry, Faculty of Pharmaceutical Science, USP, Ribeirão Preto, Brazil
| | - Isis do Carmo Kettelhut
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Laurie J Goodyear
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center, OCRC, IB, UNICAMP, Campinas, Brazil.
| |
Collapse
|
53
|
Hou Y, Su L, Zhao Y, Liu C, Yao D, Zhang M, Zhao L, Jin Y. Effect of chronic AICAR treatment on muscle fiber composition and enzyme activity in skeletal muscle of rats. JOURNAL OF APPLIED ANIMAL RESEARCH 2021. [DOI: 10.1080/09712119.2021.1889563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Yanru Hou
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Lin Su
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Yajuan Zhao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Chang Liu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Duo Yao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Min Zhang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Lihua Zhao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Ye Jin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| |
Collapse
|
54
|
Dysregulation of PGC-1α-Dependent Transcriptional Programs in Neurological and Developmental Disorders: Therapeutic Challenges and Opportunities. Cells 2021. [DOI: 10.3390/cells10020352
expr 820281011 + 880698691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Substantial evidence indicates that mitochondrial impairment contributes to neuronal dysfunction and vulnerability in disease states, leading investigators to propose that the enhancement of mitochondrial function should be considered a strategy for neuroprotection. However, multiple attempts to improve mitochondrial function have failed to impact disease progression, suggesting that the biology underlying the normal regulation of mitochondrial pathways in neurons, and its dysfunction in disease, is more complex than initially thought. Here, we present the proteins and associated pathways involved in the transcriptional regulation of nuclear-encoded genes for mitochondrial function, with a focus on the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1α). We highlight PGC-1α’s roles in neuronal and non-neuronal cell types and discuss evidence for the dysregulation of PGC-1α-dependent pathways in Huntington’s Disease, Parkinson’s Disease, and developmental disorders, emphasizing the relationship between disease-specific cellular vulnerability and cell-type-specific patterns of PGC-1α expression. Finally, we discuss the challenges inherent to therapeutic targeting of PGC-1α-related transcriptional programs, considering the roles for neuron-enriched transcriptional coactivators in co-regulating mitochondrial and synaptic genes. This information will provide novel insights into the unique aspects of transcriptional regulation of mitochondrial function in neurons and the opportunities for therapeutic targeting of transcriptional pathways for neuroprotection.
Collapse
|
55
|
Dysregulation of PGC-1α-Dependent Transcriptional Programs in Neurological and Developmental Disorders: Therapeutic Challenges and Opportunities. Cells 2021; 10:cells10020352. [PMID: 33572179 PMCID: PMC7915819 DOI: 10.3390/cells10020352] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/03/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
Substantial evidence indicates that mitochondrial impairment contributes to neuronal dysfunction and vulnerability in disease states, leading investigators to propose that the enhancement of mitochondrial function should be considered a strategy for neuroprotection. However, multiple attempts to improve mitochondrial function have failed to impact disease progression, suggesting that the biology underlying the normal regulation of mitochondrial pathways in neurons, and its dysfunction in disease, is more complex than initially thought. Here, we present the proteins and associated pathways involved in the transcriptional regulation of nuclear-encoded genes for mitochondrial function, with a focus on the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1α). We highlight PGC-1α's roles in neuronal and non-neuronal cell types and discuss evidence for the dysregulation of PGC-1α-dependent pathways in Huntington's Disease, Parkinson's Disease, and developmental disorders, emphasizing the relationship between disease-specific cellular vulnerability and cell-type-specific patterns of PGC-1α expression. Finally, we discuss the challenges inherent to therapeutic targeting of PGC-1α-related transcriptional programs, considering the roles for neuron-enriched transcriptional coactivators in co-regulating mitochondrial and synaptic genes. This information will provide novel insights into the unique aspects of transcriptional regulation of mitochondrial function in neurons and the opportunities for therapeutic targeting of transcriptional pathways for neuroprotection.
Collapse
|
56
|
D'Amico D, Marino Gammazza A, Macaluso F, Paladino L, Scalia F, Spinoso G, Dimauro I, Caporossi D, Cappello F, Di Felice V, Barone R. Sex-based differences after a single bout of exercise on PGC1α isoforms in skeletal muscle: A pilot study. FASEB J 2021; 35:e21328. [PMID: 33433932 DOI: 10.1096/fj.202002173r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/01/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022]
Abstract
To date, there are limited and incomplete data on possible sex-based differences in fiber-types of skeletal muscle and their response to physical exercise. Adult healthy male and female mice completed a single bout of endurance exercise to examine the sex-based differences of the peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α), heat shock protein 60 (Hsp60), interleukin 6 (IL-6) expression, as well as the Myosin Heavy Chain (MHC) fiber-type distribution in soleus and extensor digitorum longus (EDL) muscles. Our results showed for the first time that in male soleus, a muscle rich of type IIa fibers, endurance exercise activates specifically genes involved in mitochondrial biogenesis such as PGC1 α1 isoform, Hsp60 and IL-6, whereas the expression of PGC1 α2 and α3 was significantly upregulated in EDL muscle, a fast-twitch skeletal muscle, independently from the gender. Moreover, we found that the acute response of different PGC1α isoforms was muscle and gender dependent. These findings add a new piece to the huge puzzle of muscle response to physical exercise. Given the importance of these genes in the physiological response of the muscle to exercise, we strongly believe that our data could support future research studies to personalize a specific and sex-based exercise training protocol.
Collapse
Affiliation(s)
- Daniela D'Amico
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy.,Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Filippo Macaluso
- SMART Engineering Solutions & Technologies Research Center, eCampus University, Novedrate, Italy.,Euro-Mediterranean Institutes of Science and Technology (IEMEST), Palermo, Italy
| | - Letizia Paladino
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Federica Scalia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy.,Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, USA.,SMART Engineering Solutions & Technologies Research Center, eCampus University, Novedrate, Italy.,Euro-Mediterranean Institutes of Science and Technology (IEMEST), Palermo, Italy
| | - Giulio Spinoso
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Ivan Dimauro
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Rome, Italy
| | - Daniela Caporossi
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Rome, Italy
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy.,Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, USA.,SMART Engineering Solutions & Technologies Research Center, eCampus University, Novedrate, Italy.,Euro-Mediterranean Institutes of Science and Technology (IEMEST), Palermo, Italy
| | - Valentina Di Felice
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Rosario Barone
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| |
Collapse
|
57
|
Jevtovic F. Combination of Metformin and Exercise in Management of Metabolic Abnormalities Observed in Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2021; 14:4043-4057. [PMID: 34557007 PMCID: PMC8453852 DOI: 10.2147/dmso.s328694] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Excess nutrient intake and lack of exercise characterize the problem of obesity and are common factors in insulin resistance (IR). With an increasing number of prediabetic, and type 2 diabetic populations, metformin is still the most prescribed glucose-lowering drug and is often accompanied by recommendations for regular physical exercise. Metformin, by the inhibition of complex 1 of the electron transport chain, and exercise, by increasing energy expenditure, both elicit a low cellular energy state that leads to improvements in glucose control via activation of adenosine 5' monophosphate-activated protein kinase (AMPK). An augmented stimulation of the energy-sensing enzyme AMPK by either of the two modalities leads to an increase in glycogenolysis, glucose uptake, fat oxidation, a decrease in glycogen and protein synthesis, and gluconeogenesis in muscle and the liver, which are remarked as having positive effects on metabolic pathophysiology observed in IR and type 2 diabetes mellitus (T2DM). While both modalities exploit the energy-sensing enzyme AMPK to attain glucose homeostasis, the synergistic effect of these two treatments is not distinctly supported by the literature. Further, an antagonistic dynamic has been observed in cases where metformin and exercise were combined. Reduction of insulin-sensitizing effects of exercise and an overall hindrance of exercise performance and adaptations have been reported and could suggest the possible incongruity of these two modalities. The aim of this review is to elucidate the effect that metformin and exercise have on the management of the metabolic abnormalities observed in T2DM and to provide an insight into the interaction of these two modalities.
Collapse
Affiliation(s)
- Filip Jevtovic
- Department of Kinesiology, College of Health and Human Performance, East Carolina University, Greenville, NC, USA
- Correspondence: Filip Jevtovic East Carolina University; School of Dental Medicine, Ledyard E. Ross Hall; 1851 MacGregor Downs Road, Mail Stop 701, Greenville, NC, 27834, USATel +1 616 844 8323Fax +1 252 737 7024 Email
| |
Collapse
|
58
|
Liu J, Deng K, Pan M, Liu G, Wu J, Yang M, Huang D, Zhang W, Mai K. Dietary carbohydrates influence muscle texture of olive flounder Paralichthys olivaceus through impacting mitochondria function and metabolism of glycogen and protein. Sci Rep 2020; 10:21811. [PMID: 33311521 PMCID: PMC7732841 DOI: 10.1038/s41598-020-76255-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 10/14/2020] [Indexed: 12/13/2022] Open
Abstract
The present study was conducted to estimate the effects of dietary carbohydrates on muscle quality and the underlying mechanisms. Six isonitrogenous and isolipidic diets were formulated to contain graded levels of carbohydrates (0%, 8%, 12%, 16%, 20% and 24%, respectively). These diets were named as C0, C8, C12, C16, C20 and C24, respectively. After a 10-week feeding trial, results showed that the muscle pH, liquid holding capacity (LHC) and hardness were significantly decreased by the increasing dietary carbohydrate levels. Dietary carbohydrates significantly decreased the muscle fibre diameter, and the highest value was found in the C0 group. Accumulated glycogen and degenerated mitochondrial cristae were observed in the C24 group. Significantly higher contents of protein carbonyls were observed in the C20 group and C24 group (P < 0.05). There was a significant decrease of mtDNA copy number in the C24 group compared with that in the C0 and C8 groups. The AMP/ATP ratio in muscle decreased first and then increased with the increasing dietary carbohydrate levels. The dietary incorporation of carbohydrate significantly reduced the expression of opa1, pygm and genes involved in myogenesis (myf5 and myog). Meanwhile, proteolysis-related genes (murf-1, mafbx, capn2 and ctsl), pro-inflammatory cytokines (il-6 and tnf-α) and mstn were significantly up-regulated. In the C24 group, significant increase of phosphorylation of AMPK (Thr172), up-regulation of PGC-1α and GLUT4 were observed, while the phosphorylation level of S6 (Ser235/236) was significantly decreased. It was concluded that excessive dietary carbohydrate level (24%) had negative impacts on mitochondria function and promoted glycogen accumulation, and thereafter influenced the muscle quality of olive flounder. The activation of AMPK as well as the upregulation of PGC-1α and GLUT4 was the key mechanism.
Collapse
Affiliation(s)
- Jiahuan Liu
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China
| | - Kangyu Deng
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China
- Shenzhen Alpha Group Co., Ltd., Shenzhen, China
| | - Mingzhu Pan
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China
| | - Guangxia Liu
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China
| | - Jing Wu
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China
| | - Mengxi Yang
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China
| | - Dong Huang
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China
| | - Wenbing Zhang
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Wen Hai Road, Qingdao, 266237, China.
| | - Kangsen Mai
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Wen Hai Road, Qingdao, 266237, China
| |
Collapse
|
59
|
Naghiaee Y, Didehdar R, Pourrajab F, Rahmanian M, Heiranizadeh N, Mohiti A, Mohiti-Ardakani J. Metformin downregulates miR223 expression in insulin-resistant 3T3L1 cells and human diabetic adipose tissue. Endocrine 2020; 70:498-508. [PMID: 32970287 DOI: 10.1007/s12020-020-02459-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023]
Abstract
AIMS AND DESIGNS Metformin, an anti-diabetic drug, is the first line medication for the treatment of type 2 diabetes mellitus and some studies show its relationship with micro-RNAs. This study set up to determine the effect of metformin on miR223 expression and content of AKT/GLUT4 proteins in insulin resistant signaling in 3T3L1 cells and adipocyte of human diabetic patients. MATERIALS AND METHODS Subcutaneous adipose tissues were taken from newly diagnosed diabetic patients (HOMA-IR > 1.8), before and after three months treatment with 500 mg of metformin twice a day. Cellular homogenate was prepared and miR223 expression and AKT/GLUT4 protein expression were determined by quantitative real-time PCR and western blotting. The results were compared to insulin resistant 3T3L1 adipocytes that were treated with 10 mM Metformin. RESULTS MiR223 expression was significantly overexpressed both in insulin-resistant 3T3L1 adipocytes compared to non-insulin resistant adipocytes and in human diabetic adipose tissue, compared to non-diabetics (P value < 0.01). Metformin treatment downregulated miR223 expression in both adipocytes and human diabetic adipose tissue. In contrast the IRS/PI3-K/AKT pathway signaling components, Akt and GLUT4 increased in insulin-resistant 3T3L1 adipocytes and human diabetic adipose tissue after three months of metformin treatment. CONCLUSIONS Metformin reduced insulin resistance in adipocytes by reduction of miR223 expression and improving of IRS/Akt/GLUT4 signaling pathways. Plasma miR223 expression of human diabetic patients was reduced by metformin treatment. These results point to a novel mechanism of miR223 in insulin resistance.
Collapse
Affiliation(s)
- Yousof Naghiaee
- Department of Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Reza Didehdar
- Department of Biochemistry, Faculty of Medicine, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Pourrajab
- Department of Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Rahmanian
- Department of Endocrinology, School of Medicine Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Naeime Heiranizadeh
- Department of General Surgery, School of Medicine Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Azra Mohiti
- Department of Oral Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Mohiti-Ardakani
- Department of Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
60
|
Wang T, Wang J, Hu X, Huang XJ, Chen GX. Current understanding of glucose transporter 4 expression and functional mechanisms. World J Biol Chem 2020; 11:76-98. [PMID: 33274014 PMCID: PMC7672939 DOI: 10.4331/wjbc.v11.i3.76] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/22/2020] [Accepted: 09/22/2020] [Indexed: 02/05/2023] Open
Abstract
Glucose is used aerobically and anaerobically to generate energy for cells. Glucose transporters (GLUTs) are transmembrane proteins that transport glucose across the cell membrane. Insulin promotes glucose utilization in part through promoting glucose entry into the skeletal and adipose tissues. This has been thought to be achieved through insulin-induced GLUT4 translocation from intracellular compartments to the cell membrane, which increases the overall rate of glucose flux into a cell. The insulin-induced GLUT4 translocation has been investigated extensively. Recently, significant progress has been made in our understanding of GLUT4 expression and translocation. Here, we summarized the methods and reagents used to determine the expression levels of Slc2a4 mRNA and GLUT4 protein, and GLUT4 translocation in the skeletal muscle, adipose tissues, heart and brain. Overall, a variety of methods such real-time polymerase chain reaction, immunohistochemistry, fluorescence microscopy, fusion proteins, stable cell line and transgenic animals have been used to answer particular questions related to GLUT4 system and insulin action. It seems that insulin-induced GLUT4 translocation can be observed in the heart and brain in addition to the skeletal muscle and adipocytes. Hormones other than insulin can induce GLUT4 translocation. Clearly, more studies of GLUT4 are warranted in the future to advance of our understanding of glucose homeostasis.
Collapse
Affiliation(s)
- Tiannan Wang
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, United States
| | - Jing Wang
- College of Pharmacy, South-Central University for Nationalities, Wuhan 430074, Hubei Province, China
| | - Xinge Hu
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, United States
| | - Xian-Ju Huang
- College of Pharmacy, South-Central University for Nationalities, Wuhan 430074, Hubei Province, China
| | - Guo-Xun Chen
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, United States
| |
Collapse
|
61
|
Swim therapy-induced tissue specific metabolic responses in male rats. Life Sci 2020; 262:118516. [PMID: 33011220 DOI: 10.1016/j.lfs.2020.118516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 01/03/2023]
Abstract
Swim therapy in the form of moderate physical activity has general health benefits. Regular exercise prevents the progression of chronic diseases affecting the different bodily systems. The metabolic alterations associated with following such lifestyle remain not fully understood. The aim of the present study was to elucidate the metabolic changes following prolonged swim therapy. Twenty-four Sprague Dawley rats were divided into sedentary and exercise groups. Our results revealed that regular exercise significantly increased the serum levels of growth hormone (GH), glucagon and corticosterone. A reduction in the circulating levels of irisin and insulin hormones, and glucose were noticed alongside with an upregulation in the mRNA expression levels of FNDC5, PGC-1α, GLUT-4 and preptin receptors with downregulation in the expression of Enho gene in the heart of exercised rats. Liver of the exercised rats showed elevation in the transcriptional levels of Enho gene, PPARα, and preptin with reduction in the transcriptional levels of preptin receptors. Exercise induced an increase in the pancreatic mRNA of Enho gene, preptin and preptin receptors, and a reduction in FNDC5, PPARα and PGC-1α. An elevation in the gastrocnemius muscle PGC-1α mRNA expression and a decline in the soleus muscle Enho mRNA were found. Exercise diminishes the activities of SOD, CAT and GPx in the gastrocnemius muscle, liver and pancreas. Myogenin expression increased in all examined skeletal muscles. This study takes into account the complex crosstalk between different signaling pathways in skeletal muscles, heart, liver and pancreas as well as the metabolic alterations in response to regular exercise.
Collapse
|
62
|
Singh R, Chandel S, Dey D, Ghosh A, Roy S, Ravichandiran V, Ghosh D. Epigenetic modification and therapeutic targets of diabetes mellitus. Biosci Rep 2020; 40:BSR20202160. [PMID: 32815547 PMCID: PMC7494983 DOI: 10.1042/bsr20202160] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
The prevalence of diabetes and its related complications are increasing significantly globally. Collected evidence suggested that several genetic and environmental factors contribute to diabetes mellitus. Associated complications such as retinopathy, neuropathy, nephropathy and other cardiovascular complications are a direct result of diabetes. Epigenetic factors include deoxyribonucleic acid (DNA) methylation and histone post-translational modifications. These factors are directly related with pathological factors such as oxidative stress, generation of inflammatory mediators and hyperglycemia. These result in altered gene expression and targets cells in the pathology of diabetes mellitus without specific changes in a DNA sequence. Environmental factors and malnutrition are equally responsible for epigenetic states. Accumulated evidence suggested that environmental stimuli alter the gene expression that result in epigenetic changes in chromatin. Recent studies proposed that epigenetics may include the occurrence of 'metabolic memory' found in animal studies. Further study into epigenetic mechanism might give us new vision into the pathogenesis of diabetes mellitus and related complication thus leading to the discovery of new therapeutic targets. In this review, we discuss the possible epigenetic changes and mechanism that happen in diabetes mellitus type 1 and type 2 separately. We highlight the important epigenetic and non-epigenetic therapeutic targets involved in the management of diabetes and associated complications.
Collapse
Affiliation(s)
- Rajveer Singh
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Shivani Chandel
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Dhritiman Dey
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Arijit Ghosh
- Department of Chemistry, University of Calcutta, Kolkata 700009, India
| | - Syamal Roy
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Velayutham Ravichandiran
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Dipanjan Ghosh
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| |
Collapse
|
63
|
Gerbec ZJ, Hashemi E, Nanbakhsh A, Holzhauer S, Yang C, Mei A, Tsaih SW, Lemke A, Flister MJ, Riese MJ, Thakar MS, Malarkannan S. Conditional Deletion of PGC-1α Results in Energetic and Functional Defects in NK Cells. iScience 2020; 23:101454. [PMID: 32858341 PMCID: PMC7474003 DOI: 10.1016/j.isci.2020.101454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/30/2019] [Accepted: 08/10/2020] [Indexed: 01/07/2023] Open
Abstract
During an immune response, natural killer (NK) cells activate specific metabolic pathways to meet the increased energetic and biosynthetic demands associated with effector functions. Here, we found in vivo activation of NK cells during Listeria monocytogenes infection-augmented transcription of genes encoding mitochondria-associated proteins in a manner dependent on the transcriptional coactivator PGC-1α. Using an Ncr1Cre-based conditional knockout mouse, we found that PGC-1α was crucial for optimal NK cell effector functions and bioenergetics, as the deletion of PGC-1α was associated with decreased cytotoxic potential and cytokine production along with altered ADP/ATP ratios. Lack of PGC-1α also significantly impaired the ability of NK cells to control B16F10 tumor growth in vivo, and subsequent gene expression analysis showed that PGC-1α mediates transcription required to maintain mitochondrial activity within the tumor microenvironment. Together, these data suggest that PGC-1α-dependent transcription of specific target genes is required for optimal NK cell function during the response to infection or tumor growth.
Collapse
Affiliation(s)
- Zachary J. Gerbec
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arash Nanbakhsh
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Sandra Holzhauer
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Chao Yang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ao Mei
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shirng-Wern Tsaih
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Angela Lemke
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael J. Flister
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew J. Riese
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Monica S. Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
64
|
Santos JL, Krause BJ, Cataldo LR, Vega J, Salas-Pérez F, Mennickent P, Gallegos R, Milagro FI, Prieto-Hontoria P, Riezu-Boj JI, Bravo C, Salas-Huetos A, Arpón A, Galgani JE, Martínez JA. PPARGC1A Gene Promoter Methylation as a Biomarker of Insulin Secretion and Sensitivity in Response to Glucose Challenges. Nutrients 2020; 12:nu12092790. [PMID: 32933059 PMCID: PMC7551463 DOI: 10.3390/nu12092790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022] Open
Abstract
Methylation in CpG sites of the PPARGC1A gene (encoding PGC1-α) has been associated with adiposity, insulin secretion/sensitivity indexes and type 2 diabetes. We assessed the association between the methylation profile of the PPARGC1A gene promoter gene in leukocytes with insulin secretion/sensitivity indexes in normoglycemic women. A standard oral glucose tolerance test (OGTT) and an abbreviated version of the intravenous glucose tolerance test (IVGTT) were carried out in n = 57 Chilean nondiabetic women with measurements of plasma glucose, insulin, and C-peptide. Bisulfite-treated DNA from leukocytes was evaluated for methylation levels in six CpG sites of the proximal promoter of the PPARGC1A gene by pyrosequencing (positions -816, -783, -652, -617, -521 and -515). A strong correlation between the DNA methylation percentage of different CpG sites of the PPARGC1A promoter in leukocytes was found, suggesting an integrated epigenetic control of this region. We found a positive association between the methylation levels of the CpG site -783 with the insulin sensitivity Matsuda composite index (rho = 0.31; p = 0.02) derived from the OGTT. The CpG hypomethylation in the promoter position -783 of the PPARGC1A gene in leukocytes may represent a biomarker of reduced insulin sensitivity after the ingestion of glucose.
Collapse
Affiliation(s)
- José L. Santos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (L.R.C.); (J.V.); (P.M.); (R.G.); (C.B.); (J.E.G.)
- Correspondence: ; Tel.: +56-2-354-3862; Fax: +56-2-633-8298
| | - Bernardo J. Krause
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Avenida Libertador Bernardo O’Higgins 611, Rancagua 2841935, Chile; (B.J.K.); (F.S.-P.)
| | - Luis Rodrigo Cataldo
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (L.R.C.); (J.V.); (P.M.); (R.G.); (C.B.); (J.E.G.)
| | - Javier Vega
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (L.R.C.); (J.V.); (P.M.); (R.G.); (C.B.); (J.E.G.)
| | - Francisca Salas-Pérez
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Avenida Libertador Bernardo O’Higgins 611, Rancagua 2841935, Chile; (B.J.K.); (F.S.-P.)
- Department of Nutrition, Food Sciences and Physiology, Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain; (F.I.M.); (J.I.R.-B.); (A.A.); (J.A.M.)
| | - Paula Mennickent
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (L.R.C.); (J.V.); (P.M.); (R.G.); (C.B.); (J.E.G.)
| | - Raúl Gallegos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (L.R.C.); (J.V.); (P.M.); (R.G.); (C.B.); (J.E.G.)
| | - Fermín I. Milagro
- Department of Nutrition, Food Sciences and Physiology, Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain; (F.I.M.); (J.I.R.-B.); (A.A.); (J.A.M.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- IdiSNA, Navarra’s Health Research Institute, 31008 Pamplona, Spain
| | | | - J. Ignacio Riezu-Boj
- Department of Nutrition, Food Sciences and Physiology, Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain; (F.I.M.); (J.I.R.-B.); (A.A.); (J.A.M.)
- IdiSNA, Navarra’s Health Research Institute, 31008 Pamplona, Spain
| | - Carolina Bravo
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (L.R.C.); (J.V.); (P.M.); (R.G.); (C.B.); (J.E.G.)
| | - Albert Salas-Huetos
- Andrology and IVF Laboratory, Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA;
| | - Ana Arpón
- Department of Nutrition, Food Sciences and Physiology, Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain; (F.I.M.); (J.I.R.-B.); (A.A.); (J.A.M.)
| | - José E. Galgani
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (L.R.C.); (J.V.); (P.M.); (R.G.); (C.B.); (J.E.G.)
- Departamento de Ciencias de la Salud, Nutrición y Dietética, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 781000, Chile
| | - J. Alfredo Martínez
- Department of Nutrition, Food Sciences and Physiology, Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain; (F.I.M.); (J.I.R.-B.); (A.A.); (J.A.M.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- IdiSNA, Navarra’s Health Research Institute, 31008 Pamplona, Spain
- IMDEA-Food, 28049 Madrid, Spain
| |
Collapse
|
65
|
Chuluun-Erdene A, Sengeragchaa O, Altangerel TA, Sanjmyatav P, Dagdan B, Battulga S, Enkhbat L, Byambasuren N, Malchinkhuu M, Janlav M. Association of Candidate Gene Polymorphism with Metabolic Syndrome among Mongolian Subjects: A Case-Control Study. Med Sci (Basel) 2020; 8:medsci8030038. [PMID: 32887252 PMCID: PMC7563398 DOI: 10.3390/medsci8030038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022] Open
Abstract
Metabolic syndrome (MetS) is complex and determined by the interaction between genetic and environmental factors and their influence on obesity, insulin resistance, and related traits associated with diabetes and cardiovascular disease risk. Some dynamic markers, including adiponectin (ADIPOQ), brain-derived neurotrophic factor (BDNF), and lipoprotein lipase (LPL), are implicated in MetS; however, the influence of their genetic variants on MetS susceptibility varies in racial and ethnic groups. We investigated the association of single nucleotide polymorphism (SNP)-SNP interactions among nine SNPs in six genes with MetS's genetic predisposition in Mongolian subjects. A total of 160 patients with MetS for the case group and 144 healthy individuals for the control group were selected to participate in this study. Regression analysis of individual SNPs showed that the ADIPOQ + 45GG (odds ratio (OR) = 2.09, p = 0.011) and P+P+ of LPL PvuII (OR = 2.10, p = 0.038) carriers had an increased risk of MetS. Conversely, G allele of LPL S447X (OR = 0.45, p = 0.036) and PGC-1α 482Ser (OR = 0.26, p = 0.001) allele were estimated as protective factors, respectively. Moreover, a haplotype containing the G-P+-G combination was related to MetS. Significant loci were also related to body mass index (BMI), systolic blood pressure (SBP), serum high-density lipoprotein cholesterol (HDL-C), triglyceride (TG), and fasting blood glucose (FBG), adipokines, and insulin as well as insulin resistance (p < 0.05). Our results confirm that ADIPOQ + 45T > G, LPL PvII, and PGC-1α Gly482Ser loci are associated with MetS in Mongolian subjects.
Collapse
Affiliation(s)
- Ariunbold Chuluun-Erdene
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Orgil Sengeragchaa
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Tsend-Ayush Altangerel
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Purevjal Sanjmyatav
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Batnaran Dagdan
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
- Coronary Care Unit, Cardiovascular Center, The Shastin Central Hospital, Ulaanbaatar 16081, Mongolia
| | - Solongo Battulga
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Lundiamaa Enkhbat
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Nyamjav Byambasuren
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
| | - Munkhzol Malchinkhuu
- Department of Pathophysiology, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia;
| | - Munkhtstetseg Janlav
- Department of Biochemistry, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14210, Mongolia; (A.C.-E.); (O.S.); (T.-A.A.); (P.S.); (B.D.); (S.B.); (L.E.); (N.B.)
- Correspondence: ; Tel.: +976-9909-2287
| |
Collapse
|
66
|
McGee SL, Hargreaves M. Exercise adaptations: molecular mechanisms and potential targets for therapeutic benefit. Nat Rev Endocrinol 2020; 16:495-505. [PMID: 32632275 DOI: 10.1038/s41574-020-0377-1] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/29/2020] [Indexed: 12/19/2022]
Abstract
Exercise is fundamental for good health, whereas physical inactivity underpins many chronic diseases of modern society. It is well appreciated that regular exercise improves metabolism and the metabolic phenotype in a number of tissues. The phenotypic alterations observed in skeletal muscle are partly mediated by transcriptional responses that occur following each individual bout of exercise. This adaptive response increases oxidative capacity and influences the function of myokines and extracellular vesicles that signal to other tissues. Our understanding of the epigenetic and transcriptional mechanisms that mediate the skeletal muscle gene expression response to exercise as well as of their upstream signalling pathways has advanced substantially in the past 10 years. With this knowledge also comes the opportunity to design new therapeutic strategies based on the biology of exercise for a variety of chronic conditions where regular exercise might be a challenge. This Review provides an overview of the beneficial adaptive responses to exercise and details the molecular mechanisms involved. The possibility of designing therapeutic interventions based on these molecular mechanisms is addressed, using relevant examples that have exploited this approach.
Collapse
Affiliation(s)
- Sean L McGee
- Metabolic Research Unit, School of Medicine and Institute for Mental and Physical Health and Clinical Translation (iMPACT), Deakin University, Geelong, Victoria, Australia.
| | - Mark Hargreaves
- Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
67
|
Yvert T, Miyamoto-Mikami E, Tobina T, Shiose K, Kakigi R, Tsuzuki T, Takaragawa M, Ichinoseki-Sekine N, Pérez M, Kobayashi H, Tanaka H, Naito H, Fuku N. PPARGC1A rs8192678 and NRF1 rs6949152 Polymorphisms Are Associated with Muscle Fiber Composition in Women. Genes (Basel) 2020; 11:genes11091012. [PMID: 32867330 PMCID: PMC7563119 DOI: 10.3390/genes11091012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
PPARGC1A rs8192678 G/A (Gly482Ser) and NRF1 rs6949152 A/G polymorphisms have been associated with endurance athlete status, endurance performance phenotypes, and certain health-related markers of different pathologies such as metabolic syndrome, diabetes, and dyslipidemia. We hypothesized that they could be considered interesting candidates for explaining inter-individual variations in muscle fiber composition in humans. We aimed to examine possible associations of these polymorphisms with myosin heavy-chain (MHC) isoforms as markers of muscle fiber compositions in vastus lateralis muscle in a population of 214 healthy Japanese subjects, aged between 19 and 79 years. No significant associations were found in men for any measured variables. In contrast, in women, the PPARGC1A rs8192678 A/A genotype was significantly associated with a higher proportion of MHC-I (p = 0.042) and with a lower proportion of MHC-IIx (p = 0.033), and the NRF1 rs6949152 AA genotype was significantly associated with a higher proportion of MHC-I (p = 0.008) and with a lower proportion of MHC IIx (p = 0.035). In women, the genotype scores of the modes presenting the most significant results for PPARGC1A rs8192678 G/A (Gly482Ser) and NRF1 rs6949152 A/G polymorphisms were significantly associated with MHC-I (p = 0.0007) and MHC IIx (p = 0.0016). That is, women with combined PPARGC1A A/A and NRF1 A/A genotypes presented the highest proportion of MHC-I and the lowest proportion of MHC-IIx, in contrast to women with combined PPARGC1A GG+GA and NRF1 AG+GG genotypes, who presented the lowest proportion of MHC-I and the highest proportion of MHC-IIx. Our results suggest possible associations between these polymorphisms (both individually and in combination) and the inter-individual variability observed in muscle fiber composition in women, but not in men.
Collapse
Affiliation(s)
- Thomas Yvert
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (T.Y.); (E.M.-M.); (M.T.); (N.I.-S.); (H.N.)
- Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain;
| | - Eri Miyamoto-Mikami
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (T.Y.); (E.M.-M.); (M.T.); (N.I.-S.); (H.N.)
| | - Takuro Tobina
- Faculty of Nursing and Nutrition, University of Nagasaki, Nagasaki 851-2195, Japan;
| | - Keisuke Shiose
- Faculty of Education, University of Miyazaki, Miyazaki 889-2192, Japan;
| | - Ryo Kakigi
- Faculty of Management & Information Science, Josai International University, Chiba 283-8555, Japan;
| | | | - Mizuki Takaragawa
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (T.Y.); (E.M.-M.); (M.T.); (N.I.-S.); (H.N.)
| | - Noriko Ichinoseki-Sekine
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (T.Y.); (E.M.-M.); (M.T.); (N.I.-S.); (H.N.)
- Faculty of Liberal Arts, The Open University of Japan, Chiba 261-8586, Japan
| | - Margarita Pérez
- Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain;
| | - Hiroyuki Kobayashi
- Department of General Medicine, Mito Medical Center, Tsukuba University Hospital, Ibaraki 310-0015, Japan;
| | - Hiroaki Tanaka
- Faculty of Sports and Health Science, Fukuoka University, Fukuoka 814-0180, Japan
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (T.Y.); (E.M.-M.); (M.T.); (N.I.-S.); (H.N.)
| | - Noriyuki Fuku
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan; (T.Y.); (E.M.-M.); (M.T.); (N.I.-S.); (H.N.)
- Correspondence: ; Tel.: +81-476-98-1001 (ext. 9203)
| |
Collapse
|
68
|
Xu F, Chen R, Ma C, Tang L, Wan W, You F, Chen L, Li J, Chen Z, Liang F. ELECTROACUPUNCTURE IMPROVES INSULIN SENSITIVITY IN HIGH-FAT DIET-INDUCED INSULIN RESISTANT RATS BY ACTIVATING SIRT1 AND GLUT4 IN QUADRICEPS FEMORIS. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2020; 16:280-287. [PMID: 33363647 PMCID: PMC7748239 DOI: 10.4183/aeb.2020.280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
OBJECTIVES To assess the effects of electroacupuncture (EA) at the Zusanli (ST36), Guanyuan (CV4), Zhongwan (CV12), and Fenglong (ST40) acupoints on sirtuin 1 (SIRT1) and glucose transporter type 4 (GLUT4) expression in high-fat diet (HFD)-induced insulin-resistant (IR) rats. METHODS Wistar rats were divided into normal control (NC), HFD, and HFD+EA groups. NC rats were fed a standard chow diet and did not receive EA. After being fed an HFD for eight weeks, rats in the HFD+EA group received EA at 2 Hz five times a week for eight weeks. Rats in the HFD group did not receive EA. RESULTS In HFD-induced IR rats, EA inhibited body weight increase and water intake, which were observed in HFD rats. EA had no effect on fasting blood glucose and postprandial blood sugar levels. Intraperitoneal insulin tolerance testing revealed that EA enhanced insulin sensitivity in HFD-induced IR rats. Compared with NC rats, SIRT1 and GLUT4 were downregulated in the quadriceps femoris of HFD-fed rats but were increased after eight weeks of EA stimulation. CONCLUSIONS EA enhanced HFD-induced insulin resistance by activating SIRT1 and GLUT4 in the quadriceps femoris. These results provide powerful evidence supporting the beneficial effects of EA on HFD-induced insulin resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - F. Liang
- Hubei University of Chinese Medicine, Department of Acupuncture and Moxibustion, Wuhan, China
- Wuhan, China
| |
Collapse
|
69
|
Zhao X, Li X, Shi X, Karpac J. Diet-MEF2 interactions shape lipid droplet diversification in muscle to influence Drosophila lifespan. Aging Cell 2020; 19:e13172. [PMID: 32537848 PMCID: PMC7433001 DOI: 10.1111/acel.13172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 04/05/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
The number, size, and composition of lipid droplets can be influenced by dietary changes that shift energy substrate availability. This diversification of lipid droplets can promote metabolic flexibility and shape cellular stress responses in unique tissues with distinctive metabolic roles. Using Drosophila, we uncovered a role for myocyte enhancer factor 2 (MEF2) in modulating diet-dependent lipid droplet diversification within adult striated muscle, impacting mortality rates. Muscle-specific attenuation of MEF2, whose chronic activation maintains glucose and mitochondrial homeostasis, leads to the accumulation of large, cholesterol ester-enriched intramuscular lipid droplets in response to high calorie, carbohydrate-sufficient diets. The diet-dependent accumulation of these lipid droplets also correlates with both enhanced stress protection in muscle and increases in organismal lifespan. Furthermore, MEF2 attenuation releases an antagonistic regulation of cell cycle gene expression programs, and up-regulation of Cyclin E is required for diet- and MEF2-dependent diversification of intramuscular lipid droplets. The integration of MEF2-regulated gene expression networks with dietary responses thus plays a critical role in shaping muscle metabolism and function, further influencing organismal lifespan. Together, these results highlight a potential protective role for intramuscular lipid droplets during dietary adaptation.
Collapse
Affiliation(s)
- Xiao Zhao
- Department of Molecular and Cellular MedicineTexas A&M University Health Science CenterBryanTXUSA
| | - Xiaotong Li
- Department of Molecular and Cellular MedicineTexas A&M University Health Science CenterBryanTXUSA
| | - Xiangyu Shi
- Department of Molecular and Cellular MedicineTexas A&M University Health Science CenterBryanTXUSA
| | - Jason Karpac
- Department of Molecular and Cellular MedicineTexas A&M University Health Science CenterBryanTXUSA
| |
Collapse
|
70
|
Oka SI, Sabry AD, Horiuchi AK, Cawley KM, O’Very SA, Zaitsev MA, Shankar TS, Byun J, Mukai R, Xu X, Torres NS, Kumar A, Yazawa M, Ling J, Taleb I, Saijoh Y, Drakos SG, Sadoshima J, Warren JS. Perm1 regulates cardiac energetics as a downstream target of the histone methyltransferase Smyd1. PLoS One 2020; 15:e0234913. [PMID: 32574189 PMCID: PMC7310723 DOI: 10.1371/journal.pone.0234913] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022] Open
Abstract
The transcriptional regulatory machinery in mitochondrial bioenergetics is complex and is still not completely understood. We previously demonstrated that the histone methyltransferase Smyd1 regulates mitochondrial energetics. Here, we identified Perm1 (PPARGC-1 and ESRR-induced regulator, muscle specific 1) as a downstream target of Smyd1 through RNA-seq. Chromatin immunoprecipitation assay showed that Smyd1 directly interacts with the promoter of Perm1 in the mouse heart, and this interaction was significantly reduced in mouse hearts failing due to pressure overload for 4 weeks, where Perm1 was downregulated (24.4 ± 5.9% of sham, p<0.05). Similarly, the Perm1 protein level was significantly decreased in patients with advanced heart failure (55.2 ± 13.1% of donors, p<0.05). Phenylephrine (PE)-induced hypertrophic stress in cardiomyocytes also led to downregulation of Perm1 (55.7 ± 5.7% of control, p<0.05), and adenovirus-mediated overexpression of Perm1 rescued PE-induced downregulation of estrogen-related receptor alpha (ERRα), a key transcriptional regulator of mitochondrial energetics, and its target gene, Ndufv1 (Complex I). Pathway enrichment analysis of cardiomyocytes in which Perm1 was knocked-down by siRNA (siPerm1), revealed that the most downregulated pathway was metabolism. Cell stress tests using the Seahorse XF analyzer showed that basal respiration and ATP production were significantly reduced in siPerm1 cardiomyocytes (40.7% and 23.6% of scrambled-siRNA, respectively, both p<0.05). Luciferase reporter gene assay further revealed that Perm1 dose-dependently increased the promoter activity of the ERRα gene and known target of ERRα, Ndufv1 (Complex I). Overall, our study demonstrates that Perm1 is an essential regulator of cardiac energetics through ERRα, as part of the Smyd1 regulatory network.
Collapse
Affiliation(s)
- Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States of America
| | - Amira D. Sabry
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Amanda K. Horiuchi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Keiko M. Cawley
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Sean A. O’Very
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Maria A. Zaitsev
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Thirupura S. Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States of America
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States of America
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States of America
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Natalia S. Torres
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Anil Kumar
- Metabolic Phenotyping Core Facility, University of Utah, Salt Lake City, UT, United States of America
| | - Masayuki Yazawa
- Columbia Stem Cell Initiative, Rehabilitation and Regenerative Medicine, Columbia University, New York, NY, United States of America
- Pharmacology, Columbia University, New York, NY, United States of America
| | - Jing Ling
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
| | - Iosif Taleb
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Yukio Saijoh
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, United States of America
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States of America
| | - Junco S. Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States of America
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Institute of Resource Developmental and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
71
|
Liu Y, Yang C, Feng X, Qi L, Guo J, Zhu D, Thai PN, Zhang Y, Zhang P, Sun M, Lv J, Zhang L, Xu Z, Lu X. Prenatal High-Salt Diet-Induced Metabolic Disorders via Decreasing Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1α in Adult Male Rat Offspring. Mol Nutr Food Res 2020; 64:e2000196. [PMID: 32506826 DOI: 10.1002/mnfr.202000196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/06/2020] [Indexed: 12/14/2022]
Abstract
SCOPE Although prenatal high-salt (HS) intake leads to physiological complications in the offspring, little is known regarding its effects on the offspring's glucose metabolism. Therefore, the objectives of this study are to determine the consequences of prenatal HS diet on the offspring's metabolism and to test a potential therapy. METHODS AND RESULTS Pregnant rats are fed either a normal-salt (1% NaCl) or high-salt (8% NaCl) diet during the whole pregnancy. Experiments are conducted in five-month-old male offspring. It is found that the prenatal HS diet reduced the glucose tolerance and insulin sensitivity of the offspring. Additionally, there is down-regulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (Ppargc1a/PPARGC1A) at the transcript and protein level, which leads to decreased mitochondrial biogenesis and oxidative respiration in skeletal muscle. Moreover, the down-regulation of Ppargc1a is accompanied by decreases in the expression of glucose transporter type 4 (Glut4). With endurance exercise training, these changes are mitigated, which ultimately resulted in improved insulin resistance. CONCLUSION These findings suggest that prenatal HS intake induces metabolic disorders via the decreased expression of Ppargc1a in the skeletal muscle of adult offspring, providing novel information concerning the mechanisms and early prevention of metabolic diseases of fetal origins.
Collapse
Affiliation(s)
- Yanping Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Chunli Yang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Xueqin Feng
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Linglu Qi
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Jun Guo
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Dan Zhu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Phung N Thai
- Department of Internal Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Yingying Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Pengjie Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Juanxiu Lv
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Lubo Zhang
- Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, 92324, USA
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| | - Xiyuan Lu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, 215006, China
| |
Collapse
|
72
|
Halling JF, Pilegaard H. PGC-1α-mediated regulation of mitochondrial function and physiological implications. Appl Physiol Nutr Metab 2020; 45:927-936. [PMID: 32516539 DOI: 10.1139/apnm-2020-0005] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The majority of human energy metabolism occurs in skeletal muscle mitochondria emphasizing the importance of understanding the regulation of myocellular mitochondrial function. The transcriptional co-activator peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) has been characterized as a major factor in the transcriptional control of several mitochondrial components. Thus, PGC-1α is often described as a master regulator of mitochondrial biogenesis as well as a central player in regulating the antioxidant defense. However, accumulating evidence suggests that PGC-1α is also involved in the complex regulation of mitochondrial quality beyond biogenesis, which includes mitochondrial network dynamics and autophagic removal of damaged mitochondria. In addition, mitochondrial reactive oxygen species production has been suggested to regulate skeletal muscle insulin sensitivity, which may also be influenced by PGC-1α. This review aims to highlight the current evidence for PGC-1α-mediated regulation of skeletal muscle mitochondrial function beyond the effects on mitochondrial biogenesis as well as the potential PGC-1α-related impact on insulin-stimulated glucose uptake in skeletal muscle. Novelty PGC-1α regulates mitochondrial biogenesis but also has effects on mitochondrial functions beyond biogenesis. Mitochondrial quality control mechanisms, including fission, fusion, and mitophagy, are regulated by PGC-1α. PGC-1α-mediated regulation of mitochondrial quality may affect age-related mitochondrial dysfunction and insulin sensitivity.
Collapse
Affiliation(s)
- Jens Frey Halling
- Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark.,Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Henriette Pilegaard
- Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark.,Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
73
|
Huang H, Aminian A, Hassan M, Dan O, Axelrod CL, Schauer PR, Brethauer SA, Kirwan JP. Gastric Bypass Surgery Improves the Skeletal Muscle Ceramide/S1P Ratio and Upregulates the AMPK/ SIRT1/ PGC-1α Pathway in Zucker Diabetic Fatty Rats. Obes Surg 2020; 29:2158-2165. [PMID: 30809769 DOI: 10.1007/s11695-019-03800-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE Roux-en-Y gastric bypass (RYGB) is associated with remission of type 2 diabetes. However, the cellular and molecular mechanisms remain unknown. We hypothesized that RYGB would increase peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), sirtuin-1 (SIRT1), AMPK/pAMPK, and citrate synthase (CS) protein expression and decrease insulin resistance and these changes would be mediated by sphingolipids, including ceramides and the sphingolipid metabolite sphingosine-1 phosphate (S1P). MATERIALS AND METHODS Male ZDF rats were randomized to RYGB (n = 7) or sham surgery (n = 7) and harvested after 28 days. Total tissue ceramide, ceramide subspecies (C14:0, C16:0, C18:0, C18:1, C20:0, C24:0, and C24:1), and S1P were quantified in the white gastrocnemius muscle using LC-ESI-MS/MS after separation with HPLC. Total SIRT1, AMPK, PGC-1α, and CS protein expression were measured by Western blot. RESULTS Body weight, fasting glucose, insulin, and HOMA-IR decreased significantly after RYGB compared with sham control. These changes were paralleled by lower total ceramide (483.7 ± 32.3 vs. 280.1 ± 38.8 nmol/g wwt), C18:0 ceramide subspecies (P < 0.05), higher S1P (0.83 ± 0.05 vs. 1.54 ± 0.21 nmol/g wwt, P < 0.05), and a lower ceramide/S1P ratio (P < 0.05) in the RYGB versus sham group. AMPK, pAMPK, SIRT1, PGC-1α, and CS protein expression was also higher after RYGB (P < 0.05). The ceramide/S1P ratio correlated with weight loss (r = 0.48, P = 0.08), insulin resistance (r = 0.61, P = 0.02), PGC-1α (r = - 0.51, P < 0.06), CS (r = - 0.63, P = 0.01), and SIRT1 (r = - 0.54, P < 0.04). CONCLUSION Our data demonstrate that sphingolipid balance, and increased AMPK, SIRT1, PGC-1α, and CS protein expression are part of the mechanism that contributes to the remission of diabetes after RYGB surgery.
Collapse
Affiliation(s)
- Hazel Huang
- Department of Pathobiology, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, USA
| | - Ali Aminian
- Cleveland Clinic, Bariatric and Metabolic Institute, Cleveland, OH, USA
| | - Monique Hassan
- Cleveland Clinic, Bariatric and Metabolic Institute, Cleveland, OH, USA
| | - Olivia Dan
- Cleveland Clinic, Bariatric and Metabolic Institute, Cleveland, OH, USA
| | - Christopher L Axelrod
- Department of Pathobiology, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, USA.,Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, L-4030, Baton Rouge, LA, 70808, USA
| | - Philip R Schauer
- Cleveland Clinic, Bariatric and Metabolic Institute, Cleveland, OH, USA
| | - Stacy A Brethauer
- Cleveland Clinic, Bariatric and Metabolic Institute, Cleveland, OH, USA
| | - John P Kirwan
- Department of Pathobiology, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, USA. .,Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, L-4030, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
74
|
Miyashita K, Beppu F, Hosokawa M, Liu X, Wang S. Nutraceutical characteristics of the brown seaweed carotenoid fucoxanthin. Arch Biochem Biophys 2020; 686:108364. [PMID: 32315653 DOI: 10.1016/j.abb.2020.108364] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/10/2020] [Accepted: 04/12/2020] [Indexed: 02/08/2023]
Abstract
Fucoxanthin (Fx), a major carotenoid found in brown seaweed, is known to show a unique and wide variety of biological activities. Upon absorption, Fx is metabolized to fucoxanthinol and amarouciaxanthin, and these metabolites mainly accumulate in visceral white adipose tissue (WAT). As seen in other carotenoids, Fx can quench singlet oxygen and scavenge a wide range of free radicals. The antioxidant activity is related to the neuroprotective, photoprotective, and hepatoprotective effects of Fx. Fx is also reported to show anti-cancer activity through the regulation of several biomolecules and signaling pathways that are involved in either cell cycle arrest, apoptosis, or metastasis suppression. Among the biological activities of Fx, anti-obesity is the most well-studied and most promising effect. This effect is primarily based on the upregulation of thermogenesis by uncoupling protein 1 expression and the increase in the metabolic rate induced by mitochondrial activation. In addition, Fx shows anti-diabetic effects by improving insulin resistance and promoting glucose utilization in skeletal muscle.
Collapse
Affiliation(s)
- Kazuo Miyashita
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate, 041-8611, Japan.
| | - Fumiaki Beppu
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate, 041-8611, Japan
| | - Masashi Hosokawa
- Faculty of Fisheries Sciences, Hokkaido University, Hakodate, 041-8611, Japan
| | - Xiaoyong Liu
- Shandong Haizhibao Ocean Science and Technology Co., Ltd., Rongcheng City, 264300, China
| | - Shuzhou Wang
- Shandong Haizhibao Ocean Science and Technology Co., Ltd., Rongcheng City, 264300, China
| |
Collapse
|
75
|
Thach TT, Wu C, Hwang KY, Lee SJ. Azelaic Acid Induces Mitochondrial Biogenesis in Skeletal Muscle by Activation of Olfactory Receptor 544. Front Physiol 2020; 11:329. [PMID: 32411005 PMCID: PMC7199515 DOI: 10.3389/fphys.2020.00329] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/20/2020] [Indexed: 12/31/2022] Open
Abstract
Mouse olfactory receptor 544 (Olfr544) is ectopically expressed in varied extra-nasal organs with tissue specific functions. Here, we investigated the functionality of Olfr544 in skeletal muscle cells and tissue. The expression of Olfr544 is confirmed by RT-PCR and qPCR in skeletal muscle cells and mouse skeletal muscle assessed by RT-PCR and qPCR. Olfr544 activation by its ligand, azelaic acid (AzA, 50 μM), induced mitochondrial biogenesis and autophagy in cultured skeletal myotubes by induction of cyclic adenosine monophosphate-response element binding protein (CREB)-peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)-extracellular signal-regulated kinase-1/2 (ERK1/2) signaling axis. The silencing Olfr544 gene expression abrogated these effects of AzA in cultured myotubes. Similarly, in mice, the acute subcutaneous injection of AzA induced the CREB-PGC-1α-ERK1/2 pathways in mouse skeletal muscle, but these activations were negated in those of Olfr544 knockout mice. These demonstrate that the induction of mitochondrial biogenesis in skeletal muscle by AzA is Olfr544-dependent. Oral administration of AzA to high-fat-diet fed obese mice for 6 weeks increased mitochondrial DNA content in the skeletal muscle as well. Collectively, these findings demonstrate that Olfr544 activation by AzA regulates mitochondrial biogenesis in skeletal muscle. Intake of AzA or food containing AzA may help to improve skeletal muscle function.
Collapse
Affiliation(s)
- Trung Thanh Thach
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21-PLUS, Korea University, Seoul, South Korea.,Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Chunyan Wu
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21-PLUS, Korea University, Seoul, South Korea.,Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Kwang Yeon Hwang
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Sung-Joon Lee
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21-PLUS, Korea University, Seoul, South Korea.,Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| |
Collapse
|
76
|
Kondash ME, Ananthakumar A, Khodabukus A, Bursac N, Truskey GA. Glucose Uptake and Insulin Response in Tissue-engineered Human Skeletal Muscle. Tissue Eng Regen Med 2020; 17:801-813. [PMID: 32200516 DOI: 10.1007/s13770-020-00242-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Tissue-engineered muscles ("myobundles") offer a promising platform for developing a human in vitro model of healthy and diseased muscle for drug development and testing. Compared to traditional monolayer cultures, myobundles better model the three-dimensional structure of native skeletal muscle and are amenable to diverse functional measures to monitor the muscle health and drug response. Characterizing the metabolic function of human myobundles is of particular interest to enable their utilization in mechanistic studies of human metabolic diseases, identification of related drug targets, and systematic studies of drug safety and efficacy. METHODS To this end, we studied glucose uptake and insulin responsiveness in human tissue-engineered skeletal muscle myobundles in the basal state and in response to drug treatments. RESULTS In the human skeletal muscle myobundle system, insulin stimulates a 50% increase in 2-deoxyglucose (2-DG) uptake with a compiled EC50 of 0.27 ± 0.03 nM. Treatment of myobundles with 400 µM metformin increased basal 2-DG uptake 1.7-fold and caused a significant drop in twitch and tetanus contractile force along with decreased fatigue resistance. Treatment with the histone deacetylase inhibitor 4-phenylbutyrate (4-PBA) increased the magnitude of insulin response from a 1.2-fold increase in glucose uptake in the untreated state to a 1.4-fold increase after 4-PBA treatment. 4-PBA treated myobundles also exhibited increased fatigue resistance and increased twitch half-relaxation time. CONCLUSION Although tissue-engineered human myobundles exhibit a modest increase in glucose uptake in response to insulin, they recapitulate key features of in vivo insulin sensitivity and exhibit relevant drug-mediated perturbations in contractile function and glucose metabolism.
Collapse
Affiliation(s)
- Megan E Kondash
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | | | - Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
77
|
da Cruz BO, Cardozo LFMDF, Magliano DC, Stockler-Pinto MB. Nutritional strategies to modulate inflammation pathways via regulation of peroxisome proliferator-activated receptor β/δ. Nutr Rev 2020; 78:207-214. [PMID: 31584650 DOI: 10.1093/nutrit/nuz058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The peroxisome proliferator-activated receptor (PPAR) β/δ has an important role in multiple inflammatory conditions, including obesity, hypertension, cancer, cardiovascular disease, diabetes mellitus, and autoimmune diseases. PPARβ/δ forms a heterodimer with the retinoic acid receptor and binds to peroxisome proliferator response elements to initiate transcription of its target genes. PPARβ/δ is also able to suppress the activities of several transcription factors, including nuclear factor κB, and activator protein 1, thus regulating anti-inflammatory cellular responses and playing a protective role in several diseases. Recent studies have shown that nutritional compounds, including nutrients and bioactive compounds, can regulate PPARβ/δ expression. This review discusses key nutritional compounds that are known to modulate PPARβ/δ and are likely to affect human health.
Collapse
Affiliation(s)
- Beatriz O da Cruz
- B.O. da Cruz, L.F.M. de F. Cardozo, D.C. Magliano, and M.B. Stockler-Pinto are with the Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil
| | - Ludmila F M de França Cardozo
- B.O. da Cruz, L.F.M. de F. Cardozo, D.C. Magliano, and M.B. Stockler-Pinto are with the Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil
| | - D'Angelo C Magliano
- B.O. da Cruz, L.F.M. de F. Cardozo, D.C. Magliano, and M.B. Stockler-Pinto are with the Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil.,D.C. Magliano is with Laboratory of Morphological and Metabolic Analyses, Fluminense Federal University (UFF), Niterói-RJ, Brazil
| | - Milena B Stockler-Pinto
- B.O. da Cruz, L.F.M. de F. Cardozo, D.C. Magliano, and M.B. Stockler-Pinto are with the Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil.,M.B. Stockler-Pinto is with the Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói-RJ, Brazil
| |
Collapse
|
78
|
Choi EM, Suh KS, Park SY, Yun S, Chin SO, Rhee SY, Chon S. Orientin reduces the inhibitory effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on adipogenic differentiation and insulin signaling pathway in murine 3T3-L1 adipocytes. Chem Biol Interact 2020; 318:108978. [PMID: 32044341 DOI: 10.1016/j.cbi.2020.108978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/14/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) accumulates in human body, probably influencing adipocyte differentiation and causing various toxic effects, including wasting syndrome. Recently, orientin, a phenolic compound abundant in natural health products, has been shown to have antioxidant properties. We investigated the protective effects of orientin against TCDD-induced adipocyte dysfunction and its underlying mechanisms. In this study, orientin suppressed TCDD-induced loss of lipid accumulation. Orientin inhibited TCDD-driven decreases in the levels of peroxisome proliferator-activated receptor γ and adiponectin. Orientin also reduced TCDD-induced prostaglandin E2, and cytosolic phospholipase A2α levels, and increased TCDD-inhibited peroxisome proliferator-activated receptor gamma coactivator 1-alpha levels in 3T3-L1 adipocytes. TCDD reduced the levels of insulin receptor substrate 1 and glucose transporter 4, and decreased insulin-stimulated glucose uptake activity; however, orientin diminished these TCDD-induced effects. These results suggest that orientin may have beneficial effects on the prevention of TCDD-induced wasting syndrome and type II diabetes mellitus accompanied by insulin resistance.
Collapse
Affiliation(s)
- Eun Mi Choi
- Department of Endocrinology & Metabolism, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Kwang Sik Suh
- Department of Endocrinology & Metabolism, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - So Young Park
- Department of Endocrinology & Metabolism, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Endocrinology & Metabolism, Kyung Hee University Hospital, Seoul, 02447, Republic of Korea
| | - Soojin Yun
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Endocrinology & Metabolism, Kyung Hee University Hospital, Seoul, 02447, Republic of Korea
| | - Sang Ouk Chin
- Department of Endocrinology & Metabolism, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Endocrinology & Metabolism, Kyung Hee University Hospital, Seoul, 02447, Republic of Korea
| | - Sang Youl Rhee
- Department of Endocrinology & Metabolism, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Endocrinology & Metabolism, Kyung Hee University Hospital, Seoul, 02447, Republic of Korea
| | - Suk Chon
- Department of Endocrinology & Metabolism, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Endocrinology & Metabolism, Kyung Hee University Hospital, Seoul, 02447, Republic of Korea.
| |
Collapse
|
79
|
Fryklund C, Borg M, Svensson T, Schumacher S, Negoita F, Morén B, Stenkula KG. Impaired glucose transport in inguinal adipocytes after short-term high-sucrose feeding in mice. J Nutr Biochem 2020; 78:108338. [PMID: 32004930 DOI: 10.1016/j.jnutbio.2019.108338] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/14/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
Diets enriched in sucrose severely impair metabolic regulation and are associated with obesity, insulin resistance and glucose intolerance. In the current study, we investigated the effect of 4 weeks high-sucrose diet (HSD) feeding in C57BL6/J mice, with specific focus on adipocyte function. Mice fed HSD had slightly increased adipose tissue mass but displayed similar hepatic triglycerides, glucose and insulin levels, and glucose clearance capacity as chow-fed mice. Interestingly, we found adipose depot-specific differences, where both the non- and insulin-stimulated glucose transports were markedly impaired in primary adipocytes isolated from the inguinal fat depot from HSD-fed mice. This was accompanied by decreased protein levels of both GLUT4 and AS160. A similar but much less pronounced trend was observed in the retroperitoneal depot. In contrast, both GLUT4 expression and insulin-stimulated glucose uptake were preserved in adipocytes isolated from epididymal adipose tissue with HSD. Further, we found a slight shift in cell size distribution towards larger cells with HSD and a significant decrease of ACC and PGC-1α expression in the inguinal adipose tissue depot. Moreover, fructose alone was sufficient to decrease GLUT4 expression in cultured, mature adipocytes. Altogether, we demonstrate that short-term HSD feeding has deleterious impact on insulin response and glucose transport in the inguinal adipose tissue depot, specifically. These changes occur before the onset of systemic glucose dysmetabolism and therefore could provide a mechanistic link to overall impaired energy metabolism reported after prolonged HSD feeding, alone or in combination with HFD.
Collapse
Affiliation(s)
- Claes Fryklund
- Lund University, Department of Experimental Medical Science, 221 84 Lund, Sweden
| | - Madelene Borg
- Lund University, Department of Experimental Medical Science, 221 84 Lund, Sweden
| | - Tobias Svensson
- Lund University, Department of Experimental Medical Science, 221 84 Lund, Sweden
| | - Sara Schumacher
- Lund University, Department of Experimental Medical Science, 221 84 Lund, Sweden
| | - Florentina Negoita
- Lund University, Department of Experimental Medical Science, 221 84 Lund, Sweden
| | - Björn Morén
- Lund University, Department of Experimental Medical Science, 221 84 Lund, Sweden
| | - Karin G Stenkula
- Lund University, Department of Experimental Medical Science, 221 84 Lund, Sweden.
| |
Collapse
|
80
|
da Fonseca ACP, da Fonseca GP, Marchesini B, Voigt DD, Campos Junior M, Zembrzuski VM, Carneiro JRI, Nogueira Neto JF, Cabello PH, Cabello GMK. Genetic Variants in the Activation of the Brown-Like Adipocyte Pathway and the Risk for Severe Obesity. Obes Facts 2020; 13:130-143. [PMID: 32325455 PMCID: PMC7250364 DOI: 10.1159/000505666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 12/20/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Regular physical activity has an important role in energy expenditure and combats the development of obesity. During exercise, PPARGC1A is overexpressed, stimulating an increase of the expression of FNDC5. This protein is cleaved to release the hormone irisin, which activates a browning process in white adipose tissue through an increase in UCP1 expression. As a result, irisin leads to mitochondrial heat production and energy expenditure. OBJECTIVES The aim of this study was to investigate whether genetic variants in genes related to browning are associated with severe obesity and obesity-related features. This case-control study comprised 210 individuals with severe obesity (median body mass index [BMI] 45.6 [range 40.5-52.2]) and 191 normal-weight subjects (BMI 22.8 [21.1-23.9]). METHODS Genomic DNA was extracted from peripheral blood and the genotypes of the PPARGC1A(rs8192678, rs3736265, rs2970847, and rs3755863) and UCP1 (rs6536991 and rs12502572) genes were obtained using Taqman® assay. For the FNDC5 gene, screening of exons 3-5 as well as their intron-exon boundaries was performed using automatic sequencing. RESULTS Our results demonstrated that PPARGC1Ars2970847 and UCP1rs12502572 are associated with severe obesity. Furthermore, these polymorphisms influence anthropometric traits, such as BMI, body weight, and body adiposity index. Our findings also showed a dose-effect relationship between PPARGC1A rs8192678 and fasting plasma glucose. Finally, 5 rare mutations were identified in FNDC5, and 1 of these is a novel missense mutation. CONCLUSION This study shows that genetic variants in the activation of brown-like adipocyte pathway play an important role in the susceptibility to severe obesity.
Collapse
MESH Headings
- Adipocytes/physiology
- Adipocytes, Brown/physiology
- Adipose Tissue, Brown/physiology
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/physiology
- Adolescent
- Adult
- Aged
- Body Mass Index
- Case-Control Studies
- Cell Transdifferentiation/genetics
- Cross-Sectional Studies
- Energy Metabolism/genetics
- Female
- Fibronectins/genetics
- Genotype
- Humans
- Male
- Middle Aged
- Muscle, Skeletal/metabolism
- Mutation, Missense
- Obesity, Morbid/genetics
- Obesity, Morbid/metabolism
- Obesity, Morbid/physiopathology
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
- Polymorphism, Single Nucleotide
- Young Adult
Collapse
Affiliation(s)
| | | | - Bruna Marchesini
- Human Genetics Laboratory, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | | | - Mario Campos Junior
- Human Genetics Laboratory, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | | | - João Regis Ivar Carneiro
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Pedro Hernan Cabello
- Human Genetics Laboratory, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
- Human Genetics Laboratory, Grande Rio University, Rio de Janeiro, Brazil
| | | |
Collapse
|
81
|
Fujimoto BA, Young M, Carter L, Pang APS, Corley MJ, Fogelgren B, Polgar N. The exocyst complex regulates insulin-stimulated glucose uptake of skeletal muscle cells. Am J Physiol Endocrinol Metab 2019; 317:E957-E972. [PMID: 31593505 PMCID: PMC6962504 DOI: 10.1152/ajpendo.00109.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 01/16/2023]
Abstract
Skeletal muscle handles ~80-90% of the insulin-induced glucose uptake. In skeletal muscle, insulin binding to its cell surface receptor triggers redistribution of intracellular glucose transporter GLUT4 protein to the cell surface, enabling facilitated glucose uptake. In adipocytes, the eight-protein exocyst complex is an indispensable constituent in insulin-induced glucose uptake, as it is responsible for the targeted trafficking and plasma membrane-delivery of GLUT4. However, the role of the exocyst in skeletal muscle glucose uptake has never been investigated. Here we demonstrate that the exocyst is a necessary factor in insulin-induced glucose uptake in skeletal muscle cells as well. The exocyst complex colocalizes with GLUT4 storage vesicles in L6-GLUT4myc myoblasts at a basal state and associates with these vesicles during their translocation to the plasma membrane after insulin signaling. Moreover, we show that the exocyst inhibitor endosidin-2 and a heterozygous knockout of Exoc5 in skeletal myoblast cells both lead to impaired GLUT4 trafficking to the plasma membrane and hinder glucose uptake in response to an insulin stimulus. Our research is the first to establish that the exocyst complex regulates insulin-induced GLUT4 exocytosis and glucose metabolism in muscle cells. A deeper knowledge of the role of the exocyst complex in skeletal muscle tissue may help our understanding of insulin resistance in type 2 diabetes.
Collapse
Affiliation(s)
- Brent A Fujimoto
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Madison Young
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Lamar Carter
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Alina P S Pang
- Department of Native Hawaiian Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Michael J Corley
- Department of Native Hawaiian Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Ben Fogelgren
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Noemi Polgar
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| |
Collapse
|
82
|
Regulation of metabolism during hibernation in brown bears (Ursus arctos): Involvement of cortisol, PGC-1α and AMPK in adipose tissue and skeletal muscle. Comp Biochem Physiol A Mol Integr Physiol 2019; 240:110591. [PMID: 31669707 DOI: 10.1016/j.cbpa.2019.110591] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/15/2019] [Accepted: 10/18/2019] [Indexed: 12/19/2022]
Abstract
The purpose of this study was to investigate changes in expression of known cellular regulators of metabolism during hyperphagia (Sept) and hibernation (Jan) in skeletal muscle and adipose tissue of brown bears and determine whether signaling molecules and transcription factors known to respond to changes in cellular energy state are involved in the regulation of these metabolic adaptations. During hibernation, serum levels of cortisol, glycerol, and triglycerides were elevated, and protein expression and activation of AMPK in skeletal muscle and adipose tissue were reduced. mRNA expression of the co-activator PGC-1α was reduced in all tissues in hibernation whereas mRNA expression of the transcription factor PPAR-α was reduced in the vastus lateralis muscle and adipose tissue only. During hibernation, gene expression of ATGL and CD36 was not altered; however, HSL gene expression was reduced in adipose tissue. During hibernation gene expression of the lipogenic enzyme DGAT in all tissues and the expression of the FA oxidative enzyme LCAD in the vastus lateralis muscle were reduced. Gene and protein expression of the glucose transporter GLUT4 was decreased in adipose tissue in hibernation. Our data suggest that high cortisol levels are a key adaptation during hibernation and link cortisol to a reduced activation of the AMPK/PGC-1α/PPAR-α axis in the regulation of metabolism in skeletal muscle and adipose tissue. Moreover, our results indicate that during this phase of hibernation at a time when metabolic rate is significantly reduced metabolic adaptations in peripheral tissues seek to limit the detrimental effects of unduly large energy dissipation.
Collapse
|
83
|
Meta-analyses of the association between the PPARGC1A Gly482Ser polymorphism and athletic performance. Biol Sport 2019; 36:301-309. [PMID: 31938000 PMCID: PMC6945052 DOI: 10.5114/biolsport.2019.88752] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/22/2019] [Accepted: 07/03/2019] [Indexed: 01/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) encoded by the PPARGC1A gene is a vital regulator of glucose and fatty acid oxidation, mitochondrial biogenesis, and skeletal muscle fibre conversion. Several studies have investigated the association between PPARGC1A Gly482Ser polymorphism and athletic performance in humans. However, the results were contradictory. In the present study, two meta-analyses were performed to assess the association between the Gly482Ser polymorphism and endurance or power athletic performance to resolve this inconsistency. Ten articles were identified, including a total of 3,708 athletes and 6,228 controls. Higher frequencies of the Gly/Gly genotype (OR, 1.26; 95% CI, 1.11-1.42) and the Gly allele (OR, 1.29; 95% CI, 1.09-1.52) were observed in Caucasian endurance athletes. Furthermore, higher incidences of the Gly/Gly genotype (OR, 1.30; 95% CI, 1.16-1.46) and the Gly allele (OR, 1.22; 95% CI, 1.12-1.33) were observed in power athletes compared to controls. This finding demonstrates that the Gly/Gly genotype and the Gly allele of the PPARGC1A Gly482Ser polymorphism may facilitate athletic performance regardless of the type of sport, as well as providing solid evidence to support the possible influence of genetic factors on human athletic performance.
Collapse
|
84
|
PLCε regulates prostate cancer mitochondrial oxidative metabolism and migration via upregulation of Twist1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:337. [PMID: 31383001 PMCID: PMC6683382 DOI: 10.1186/s13046-019-1323-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/14/2019] [Indexed: 12/17/2022]
Abstract
Background Metabolic rewiring is a common feature of many cancer types, including prostate cancer (PCa). Alterations in master genes lead to mitochondrial metabolic rewiring and provide an appealing target to inhibit cancer progression and improve survival. Phospholipase C (PLC)ε is a regulator of tumor generation and progression. However, its role in mitochondrial metabolism remains unclear. Methods The GEO, The Cancer Genome Atlas, and the GTEx databases were used to determine Twist1 mRNA levels in tumors and their non-tumor counterparts. Fifty-five PCa and 48 benign prostatic hypertrophy tissue samples were tested for the presence of PLCε and Twist1 immunohistochemically. An association between PLCε and Twist1 was determined by Pearson’s correlation analysis. PLCε was knocked down with a lentiviral short hairpin RNA. Mitochondrial activity was assessed by measuring the oxygen consumption rate. Western blotting analyses were used to measure levels of PPARβ, Twist1, phosphorylated (p)-Twist1, p-MEK, p-ERK, p-P38, and p-c-Jun N-terminal kinase (JNK). Cells were treated with inhibitors of MEK, JNK, and P38 MAPK, and an agonist and inhibitor of peroxisome proliferator activated receptor (PPAR) β, to evaluate which signaling pathways were involved in PLCε-mediated Twist1 expression. The stability of Twist1 was determined after blocking protein synthesis with cycloheximide. Reporter assays utilized E-cadherin or N-cadherin luciferase reporters under depletion of PLCε or Twist1. Transwell assays assessed cell migration. Finally, a nude mouse tumor xenograft assay was conducted to verify the role of PLCε in tumor formation. Results Our findings revealed that the expression of PLCε was positively associated with Twist1 in clinical PCa samples. PLCε knockdown promoted mitochondrial oxidative metabolism in PCa cells. Mechanistically, PLCε increased phosphorylation of Twist1 and stabilized the Twist1 protein through MAPK signaling. The transcriptional activity of Twist1, and the Twist1-mediated epithelial-to-mesenchymal transition, cell migration, and transcription regulation, were suppressed by PLCε knockdown and by blocking PPARβ nuclear translocation. The tumor xenograft assay demonstrated that PLCε depletion diminished PCa cell tumorigenesis. Conclusions These findings reveal an undiscovered physiological role for PLCε in the suppression of mitochondrial oxidative metabolism that has significant implications for understanding PCa occurrence and migration. Electronic supplementary material The online version of this article (10.1186/s13046-019-1323-8) contains supplementary material, which is available to authorized users.
Collapse
|
85
|
Koh JH, Johnson ML, Dasari S, LeBrasseur NK, Vuckovic I, Henderson GC, Cooper SA, Manjunatha S, Ruegsegger GN, Shulman GI, Lanza IR, Nair KS. TFAM Enhances Fat Oxidation and Attenuates High-Fat Diet-Induced Insulin Resistance in Skeletal Muscle. Diabetes 2019; 68:1552-1564. [PMID: 31088855 PMCID: PMC6692815 DOI: 10.2337/db19-0088] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/05/2019] [Indexed: 12/11/2022]
Abstract
Diet-induced insulin resistance (IR) adversely affects human health and life span. We show that muscle-specific overexpression of human mitochondrial transcription factor A (TFAM) attenuates high-fat diet (HFD)-induced fat gain and IR in mice in conjunction with increased energy expenditure and reduced oxidative stress. These TFAM effects on muscle are shown to be exerted by molecular changes that are beyond its direct effect on mitochondrial DNA replication and transcription. TFAM augmented the muscle tricarboxylic acid cycle and citrate synthase facilitating energy expenditure. TFAM enhanced muscle glucose uptake despite increased fatty acid (FA) oxidation in concert with higher β-oxidation capacity to reduce the accumulation of IR-related carnitines and ceramides. TFAM also increased pAMPK expression, explaining enhanced PGC1α and PPARβ, and reversing HFD-induced GLUT4 and pAKT reductions. TFAM-induced mild uncoupling is shown to protect mitochondrial membrane potential against FA-induced uncontrolled depolarization. These coordinated changes conferred protection to TFAM mice against HFD-induced obesity and IR while reducing oxidative stress with potential translational opportunities.
Collapse
Affiliation(s)
- Jin-Ho Koh
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN
| | - Matthew L Johnson
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Nathan K LeBrasseur
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Ivan Vuckovic
- Mayo Clinic Regional Comprehensive Metabolomics Core, Mayo Clinic, Rochester, MN
| | | | - Shawna A Cooper
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN
| | | | | | - Gerald I Shulman
- Department of Medicine and Cellular and Molecular Physiology, Yale University, New Haven, CT
| | - Ian R Lanza
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
86
|
MIZUSAKI NAO, NOMURA KAZUHIRO, HOSOOKA TETSUYA, SHIOMI MASASHI, OGAWA KAZUO, TSUNODA TETSUTO, TAMORI YOSHIKAZU, OGAWA WATARU. The Novel Lipid-Lowering Drug D-47 Ameliorates Hepatic Steatosis and Promotes Brown/Beige-Like Change of White Adipose Tissue in db/db Mice. THE KOBE JOURNAL OF MEDICAL SCIENCES 2019; 65:E36-E43. [PMID: 31341155 PMCID: PMC6668594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/05/2019] [Indexed: 06/10/2023]
Abstract
D-47 is a newly developed solid dispersion of the arginine salt of (S)-(+)-4-[1-(4-tert-butylphenyl)-2-oxo-pyrrolidin-4-yl]methoxybenzoic acid (S-2E), which inhibits sterol and fatty acid synthesis. D-47 was recently shown to lower the serum level and hepatic content of both triglyceride and cholesterol in a rabbit model of familial hypercholesterolemia. We here investigated the effects of D-47 on dyslipidemia and hepatic steatosis in comparison with those of bezafibrate in the db/db mouse model of obesity. Treatment of db/db mice with D-47 or bezafibrate for 14 days lowered the serum triglyceride concentration without affecting that of cholesterol. D-47, but not bezafibrate, almost completely eliminated lipid droplets in hepatocytes and markedly lowered the triglyceride content of the liver in these mice. The two agents induced similar changes in the hepatic expression of genes including those related to β-oxidation or fatty acid synthesis. D-47 however significantly reduced the mass of white adipose tissue and up-regulated the expression of genes related to energy expenditure, mitochondrial function, fatty acid oxidation or lipolysis in this tissue, indicating that D-47 induced the brown/beige adipocyte-like change in white adipose tissue, whereas bezafibrate had no such effects. Treatment of 3T3-L1 adipocytes with D-47 provoked the expression of genes related to mitochondrial function, fatty acid oxidation or lipolysis. Our data have thus shown that D-47 ameliorated hypertriglyceridemia and hepatic steatosis in an animal model of obesity, and they suggest that this latter effect might be mediated through the change of adipose tissue characteristics.
Collapse
Affiliation(s)
- NAO MIZUSAKI
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - KAZUHIRO NOMURA
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - TETSUYA HOSOOKA
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - MASASHI SHIOMI
- Institute for Experimental Animals, Kobe University Graduate School of Medicine, Kobe, Japan
| | - KAZUO OGAWA
- Medical Chemistry, Faculty of Pharmaceutical Science, Tokushima Bunri University, Tokushima, Japan
| | - TETSUTO TSUNODA
- Medical Chemistry, Faculty of Pharmaceutical Science, Tokushima Bunri University, Tokushima, Japan
| | - YOSHIKAZU TAMORI
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Creative Health Promotion, Department of Social/Community Medicine and Health Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - WATARU OGAWA
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
87
|
Felipe-Abrio B, Verdugo-Sivianes EM, Carnero A. c-MYB- and PGC1a-dependent metabolic switch induced by MYBBP1A loss in renal cancer. Mol Oncol 2019; 13:1519-1533. [PMID: 31066170 PMCID: PMC6599841 DOI: 10.1002/1878-0261.12499] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/24/2019] [Accepted: 05/07/2019] [Indexed: 12/22/2022] Open
Abstract
The tumor microenvironment may alter the original tumorigenic potential of tumor cells. Under harsh environmental conditions, genetic alterations conferring selective advantages may initiate the growth of tumor subclones, providing new opportunities for these tumors to grow. We performed a genetic loss-of-function screen to identify genetic alterations able to promote tumor cell growth in the absence of glucose. We identified that downregulation of MYBBP1A increases tumorigenic properties under nonpermissive conditions. MYBBP1A downregulation simultaneously activates PGC1α, directly by alleviating direct repression and indirectly by increasing PGC1α mRNA levels through c-MYB, leading to a metabolic switch from glycolysis to OXPHOS and increased tumorigenesis in low-glucose microenvironments. We have also identified reduced MYBBP1A expression in human renal tumor samples, which show high expression levels of genes involved in oxidative metabolism. In summary, our data support the role of MYBBP1A as a tumor suppressor by regulating c-MYB and PGC1α. Therefore, loss of MYBBP1A increases adaptability spanning of tumors through metabolic switch.
Collapse
Affiliation(s)
- Blanca Felipe-Abrio
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Eva M Verdugo-Sivianes
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
88
|
Abstract
Inflammatory processes underlie many diseases associated with injury of the heart muscle, including conditions without an obvious inflammatory pathogenic component such as hypertensive and diabetic cardiomyopathy. Persistence of cardiac inflammation can cause irreversible structural and functional deficits. Some are induced by direct damage of the heart muscle by cellular and soluble mediators but also by metabolic adaptations sustained by the inflammatory microenvironment. It is well established that both cardiomyocytes and immune cells undergo metabolic reprogramming in the site of inflammation, which allow them to deal with decreased availability of nutrients and oxygen. However, like in cancer, competition for nutrients and increased production of signalling metabolites such as lactate initiate a metabolic cross-talk between immune cells and cardiomyocytes which, we propose, might tip the balance between resolution of the inflammation versus adverse cardiac remodeling. Here we review our current understanding of the metabolic reprogramming of both heart tissue and immune cells during inflammation, and we discuss potential key mechanisms by which these metabolic responses intersect and influence each other and ultimately define the prognosis of the inflammatory process in the heart.
Collapse
Affiliation(s)
- Federica M Marelli-Berg
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Dunja Aksentijevic
- School of Biological and Chemical Sciences, Queen Mary University of London, G.E. Fogg Building, Mile End Road, London E1 4NS, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
89
|
Abstract
The two types of thermogenic fat cells, beige and brown adipocytes, play a significant role in regulating energy homeostasis. Their development and thermogenesis are tightly regulated by dynamic epigenetic mechanisms, which could potentially be targeted to treat metabolic disorders such as obesity. However, we are just beginning to catalog and understand these dynamic changes. In this review, we will discuss the current understanding of the role of DNA (de)methylation events in beige and brown adipose biology in order to highlight the holes in our knowledge and to point the way forward for future studies.
Collapse
Affiliation(s)
- Han Xiao
- a Department of Nutritional Sciences and Toxicology, UC Berkeley , Berkeley , CA , USA
| | - Sona Kang
- a Department of Nutritional Sciences and Toxicology, UC Berkeley , Berkeley , CA , USA
| |
Collapse
|
90
|
Yuan D, Xiao D, Gao Q, Zeng L. PGC-1α activation: a therapeutic target for type 2 diabetes? Eat Weight Disord 2019; 24:385-395. [PMID: 30498989 DOI: 10.1007/s40519-018-0622-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/24/2018] [Indexed: 12/19/2022] Open
Abstract
Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) has gained popularity as a very attractive target for diabetic therapies due to its role in lipid and glucose metabolism. Pharmacological activation of PGC-1α is thought to elicit health benefits. However, this notion has been questioned by increasing evidence, which suggests that insulin resistant is exacerbated when PGC-1α expression is far beyond normal physiological limits and is prevented under the condition of PGC-1α deficiency. This narrative review suggests that PGC-1α, as a master metabolic regulator, exerts roles in insulin sensitivity in a tissue-specific manner and in a physical activity/age-dependent fashion. When using PGC-1α as a target for therapeutic strategies against insulin resistance and T2DM, we should take these factors into consideration.Level of evidence: Level V, narrative review.
Collapse
Affiliation(s)
- Daixiu Yuan
- Department of Medicine, Jishou University, Jishou, 41600, Hunan, China
| | - Dingfu Xiao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Qian Gao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Liming Zeng
- Science College of Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China.
| |
Collapse
|
91
|
Fang P, He B, Yu M, Shi M, Zhu Y, Zhang Z, Bo P. Treatment with celastrol protects against obesity through suppression of galanin-induced fat intake and activation of PGC-1α/GLUT4 axis-mediated glucose consumption. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1341-1350. [PMID: 30742994 DOI: 10.1016/j.bbadis.2019.02.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 01/14/2019] [Accepted: 02/04/2019] [Indexed: 12/27/2022]
Abstract
Overweight and obesity may cause several metabolic complications, including type 2 diabetes mellitus and hyperlipidemia. Despite years of progress in medicine, there are no highly effective pharmacological treatments for obesity. The natural compound celastrol, a pentacyclic triterpene extracted from the roots of Tripterygium Wilfordi (thunder god vine) plant, exerts various bioactivities including anti-diabetic and anti-obese effects. Although celastrol could decrease food intake and obesity, the detailed mechanism for celastrol is still unclear as yet. Herein, we intended to determine the effect of celastrol on obesity and the underlying mechanisms. In the present study, diet-induced obese mice were treated with 100 μg/kg/d celastrol for the last 21 days, and 3T3-L1 cells were treated with celastrol for 6 h. The present findings showed that celastrol suppresses fat intake, and leads to weight loss by inhibiting galanin and its receptor expression in the hypothalamus of mice fed a high-fat diet. More importantly, in addition to these direct anti-obesity activities, celastrol augmented the PGC-1α and GLUT4 expression in adipocytes and skeletal muscles to increase glucose uptake through AKT and P38 MAPK activation. Celastrol also inhibited gluconeogenic activity through a CREB/PGC-1α pathway. In conclusion, the weight-lowering effects of celastrol are driven by decreased galanin-induced food consumption. Thus, this study contributes to our understanding of the anti-obese role of celastrol, and provides a possibility of using celastrol to treat obesity in clinic.
Collapse
Affiliation(s)
- Penghua Fang
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Biao He
- College of Physical Education, Anhui Normal University, Wuhu, Anhui 241003, China
| | - Mei Yu
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Mingyi Shi
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Yan Zhu
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| | - Ping Bo
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
92
|
NURR1 activation in skeletal muscle controls systemic energy homeostasis. Proc Natl Acad Sci U S A 2019; 116:11299-11308. [PMID: 31110021 DOI: 10.1073/pnas.1902490116] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Skeletal muscle plays a central role in the control of metabolism and exercise tolerance. Analysis of muscle enhancers activated after exercise in mice revealed the orphan nuclear receptor NURR1/NR4A2 as a prominent component of exercise-responsive enhancers. We show that exercise enhances the expression of NURR1, and transgenic overexpression of NURR1 in skeletal muscle enhances physical performance in mice. NURR1 expression in skeletal muscle is also sufficient to prevent hyperglycemia and hepatic steatosis, by enhancing muscle glucose uptake and storage as glycogen. Furthermore, treatment of obese mice with putative NURR1 agonists increases energy expenditure, improves glucose tolerance, and confers a lean phenotype, mimicking the effects of exercise. These findings identify a key role for NURR1 in governance of skeletal muscle glucose metabolism, and reveal a transcriptional link between exercise and metabolism. Our findings also identify NURR1 agonists as possible exercise mimetics with the potential to ameliorate obesity and other metabolic abnormalities.
Collapse
|
93
|
Inayat H, Azim MK, Baloch AA. Analysis of Inflammatory Gene Expression Profile of Peripheral Blood Leukocytes in Type 2 Diabetes. Immunol Invest 2019; 48:618-631. [PMID: 30961396 DOI: 10.1080/08820139.2019.1586917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The pathophysiology of type 2 diabetes (T2DM) is associated with perturbation of innate immune response. Several studies indicated alteration of pro-inflammatory and anti-inflammatory cytokines, chemokines and other mediators of innate immune response in T2DM. This study was designed to perform quantitative PCR-based expression profiling of genes involved in inflammation (i.e. CASP1, CASP5, CCL5, CXC11, CCR5, NF-Κb, IL-4, PPARG and PGC1α) in peripheral blood leukocytes of T2DM patients. The T2DM patients are often prescribed with metformin and insulin while metformin has also been reported to possess anti-inflammatory activity. To address the question whether metformin exerts any effect on inflammatory mediators in bloodstream, human subjects in this study were divided into four groups on the basis of medication they were taking during last 6 month. These groups included NT-T2DM (T2DM patients not taking medication, n = 34), Met-T2DM (T2DM patients taking metformin, n = 33), INS-T2DM (T2DM patients taking insulin, n = 15) and NGT (normoglycemic subjects, n = 34) groups. Differential expression of gene transcripts at a cutoff of fourfold was considered significant. In the NT-T2DM group, transcripts of inflammation-related genes (i.e. CASP1, CASP5, CCL5, CCR5 and NF-kB) were up-regulated while transcripts of PPARG and PGC1α genes were down-regulated compared to NGT group. On the other hand, down-regulation of CASP1, CASP5, CCL5, CCR5 and NF-kB transcripts was evident in Met-T2DM and INS-T2DM groups when compared to the NT-T2DM group. The Met-T2DM group and INS-T2DM group showed a significant difference in the transcript level of CASP1 and CCL5 which are more down-regulated in the Met-T2DM group compared to INS-T2DM group. These findings indicated that (a) in T2DM, expression of inflammation-related genes is up-regulated and (b) anti-inflammatory activity of metformin appears to be independent of its anti-hyperglycemic activity.
Collapse
Affiliation(s)
- Humera Inayat
- a H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences , University of Karachi , Karachi , Pakistan
| | - M Kamran Azim
- b Department of Biosciences , Mohammad Ali Jinnah University , Karachi , Pakistan
| | - Akhter Ali Baloch
- c National Institute of Diabetes and Endocrinology , Dow University Health Sciences , Karachi , Pakistan
| |
Collapse
|
94
|
Bost F, Kaminski L. The metabolic modulator PGC-1α in cancer. Am J Cancer Res 2019; 9:198-211. [PMID: 30906622 PMCID: PMC6405967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 09/21/2018] [Indexed: 06/09/2023] Open
Abstract
The peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) is a central modulator of cell metabolism. It regulates mitochondrial biogenesis and oxidative metabolism. Modifications and adaptations in cellular metabolism are hallmarks of cancer cells, thus, it is not surprising that PGC-1α plays a role in cancer. Several recent articles have shown that PGC-1α expression is altered in tumors and metastasis in relation to modifications in cellular metabolism. The potential uses of PGC-1α as a therapeutic target and a biomarker of the advanced form of cancer will be summarized in this review.
Collapse
Affiliation(s)
- Frederic Bost
- Université Nice Côte d'Azur, Inserm, C3M, Centre Méditerranéen de Médecine Moléculaire (INSERM U1065) Nice, France
| | - Lisa Kaminski
- Université Nice Côte d'Azur, Inserm, C3M, Centre Méditerranéen de Médecine Moléculaire (INSERM U1065) Nice, France
| |
Collapse
|
95
|
Song X, Rahimnejad S, Zhou W, Cai L, Lu K. Molecular Characterization of Peroxisome Proliferator-Activated Receptor-Gamma Coactivator-1α (PGC1α) and Its Role in Mitochondrial Biogenesis in Blunt Snout Bream ( Megalobrama amblycephala). Front Physiol 2019; 9:1957. [PMID: 30733687 PMCID: PMC6354234 DOI: 10.3389/fphys.2018.01957] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 12/23/2018] [Indexed: 11/13/2022] Open
Abstract
PGC1α is a transcriptional coactivator that plays key roles in mitochondrial biogenesis, so exploring its molecular characterization contributes to the understanding of mitochondrial function in cultured fish. In the present study, a full-length cDNA coding PGC1α was cloned from the liver of blunt snout bream (Megalobrama amblycephala) which covered 3741 bp with an open reading frame of 2646 bp encoding 881 amino acids. Sequence alignment and phylogenetic analysis revealed high conservation with other fish species, as well as other higher vertebrates. Comparison of the derived amino acid sequences indicates that, as with other fish, there is a proline at position 176 (RIRP) compared to a Thr in the mammalian sequences (RIRT). To investigate PGC1α function, three in vitro tests were carried out using primary hepatocytes of blunt snout bream. The effect of AMPK activity on the expression of PGC1α was determined by the culture of the hepatocytes with an activator (Metformin) or inhibitor (Compound C) of AMPK. Neither AMPK activation nor inhibition altered PGC1α expression. Knockdown of PGC1α expression in hepatocytes using small interfering RNA (si-RNA) was used to determine the role of PGC1α in mitochondrial biogenesis. No significant differences in the expression of NRF1 and TFAM, and mtDNA copy number were found between control and si-RNA groups. Also, hepatocytes were cultured with oleic acid, and the findings showed the significant reduction of mtDNA copy number in oleic acid group compared to control. Moreover, oleic acid down-regulated the expression of NRF1 and TFAM genes, while PGC1α expression remained unchanged. Our findings support the proposal that PGC1α may not play a role in mitochondrial biogenesis in blunt snout bream hepatocytes.
Collapse
Affiliation(s)
- Xiaojun Song
- Laboratory for Animal Nutrition and Immune Molecular Biology, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Samad Rahimnejad
- Laboratory of Aquatic Animal Nutrition and Physiology, Fisheries College, Jimei University, Xiamen, China.,South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Institute of Aquaculture and Protection of Waters, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, České Budějovice, Czechia
| | - Wenhao Zhou
- Laboratory of Aquatic Animal Nutrition and Physiology, Fisheries College, Jimei University, Xiamen, China
| | - Linsen Cai
- Laboratory of Aquatic Animal Nutrition and Physiology, Fisheries College, Jimei University, Xiamen, China
| | - Kangle Lu
- Laboratory of Aquatic Animal Nutrition and Physiology, Fisheries College, Jimei University, Xiamen, China
| |
Collapse
|
96
|
Tsai CC, Tiao MM, Sheen JM, Huang LT, Tain YL, Lin IC, Lin YJ, Lai YJ, Chen CC, Chang KA, Yu HR. Obesity programmed by prenatal dexamethasone and postnatal high-fat diet leads to distinct alterations in nutrition sensory signals and circadian-clock genes in visceral adipose tissue. Lipids Health Dis 2019; 18:19. [PMID: 30658634 PMCID: PMC6339284 DOI: 10.1186/s12944-019-0963-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/06/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Prenatal dexamethasone treatment has been shown to enhance the susceptibility of offspring to postnatal high-fat (HF) diet-induced programmed obesity. We investigated the metabolic phenotypes, nutrient-sensing signal and circadian-clock genes in adipose tissue that are programmed by prenatal dexamethasone exposure and postnatal HF diet. METHODS Male offspring of Sprague-Dawley rats were divided into four experimental groups: normal diet, prenatal dexamethasone exposure, postnatal HF diet, and prenatal dexamethasone plus postnatal HF diet. Postnatal HF diet was prescribed from weaning to 6 months of age. RESULTS Prenatal dexamethasone and postnatal HF diet exerted synergistic effects on body weight and visceral adiposity, whereas prenatal dexamethasone and postnatal HF diet altered the metabolic profile and caused leptin dysregulation. Prenatal dexamethasone and postnatal HF diet distinctly influenced nutrient-sensing molecules and circadian-clock genes in adipose tissue. The mRNA expression of mTOR, AMPK-α2, PPAR-α, and PPAR-γ was suppressed by prenatal dexamethasone but enhanced by postnatal HF diet. CONCLUSION Prenatal dexamethasone and postnatal HF treatment cause dysregulation of nutrient-sensing molecules and circadian-clock genes in visceral adipose tissue. Characterizing altered nutrient-sensing molecules and circadian-clock genes has potential therapeutic relevance with respect to the pathogenesis and treatment of prenatal stress and postnatal HF diet-related metabolic disorders.
Collapse
Affiliation(s)
- Ching-Chou Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, 83301, Taiwan.,Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Mao-Meng Tiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, 123 Ta-Pei Road, Niao Sung, Kaohsiung, 83301, Taiwan, Republic of China
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, 123 Ta-Pei Road, Niao Sung, Kaohsiung, 83301, Taiwan, Republic of China
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, 123 Ta-Pei Road, Niao Sung, Kaohsiung, 83301, Taiwan, Republic of China
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, 123 Ta-Pei Road, Niao Sung, Kaohsiung, 83301, Taiwan, Republic of China
| | - I-Chun Lin
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, 123 Ta-Pei Road, Niao Sung, Kaohsiung, 83301, Taiwan, Republic of China
| | - Yu-Ju Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, 83301, Taiwan
| | - Yun-Ju Lai
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, 83301, Taiwan
| | - Chih-Cheng Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, 123 Ta-Pei Road, Niao Sung, Kaohsiung, 83301, Taiwan, Republic of China
| | - Kow-Aung Chang
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, 83301, Taiwan
| | - Hong-Ren Yu
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan. .,Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, 123 Ta-Pei Road, Niao Sung, Kaohsiung, 83301, Taiwan, Republic of China.
| |
Collapse
|
97
|
Kawabeta K, Hase-Tamaru S, Yuasa M, Suruga K, Sugano M, Koba K. Dietary β-Conglycinin Modulates Insulin Sensitivity, Body Fat Mass, and Lipid Metabolism in Obese Otsuka Long-Evans Tokushima Fatty (OLETF) Rats. J Oleo Sci 2019; 68:339-350. [DOI: 10.5650/jos.ess18232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Koji Kawabeta
- Graduate School of Human Health Science, University of Nagasaki
| | - Shizuka Hase-Tamaru
- Faculty of Nursing and Nutrition, University of Nagasaki
- Department of Life, Environment and Materials Science, Fukuoka Institute of Technology
| | - Masahiro Yuasa
- Faculty of Nursing and Nutrition, University of Nagasaki
| | - Kazuhito Suruga
- Graduate School of Human Health Science, University of Nagasaki
- Faculty of Nursing and Nutrition, University of Nagasaki
| | - Michihiro Sugano
- Professor Emeritus, Kyushu University and Prefectural University of Kumamoto
| | - Kazunori Koba
- Graduate School of Human Health Science, University of Nagasaki
- Faculty of Nursing and Nutrition, University of Nagasaki
| |
Collapse
|
98
|
Kitada M, Ogura Y, Monno I, Koya D. Sirtuins and Type 2 Diabetes: Role in Inflammation, Oxidative Stress, and Mitochondrial Function. Front Endocrinol (Lausanne) 2019; 10:187. [PMID: 30972029 PMCID: PMC6445872 DOI: 10.3389/fendo.2019.00187] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 03/06/2019] [Indexed: 01/05/2023] Open
Abstract
The rising incidence of type 2 diabetes mellitus (T2DM) is a major public health concern, and novel therapeutic strategies to prevent T2DM are urgently needed worldwide. Aging is recognized as one of the risk factors for metabolic impairments, including insulin resistance and T2DM. Inflammation, oxidative stress, and mitochondrial dysfunction are closely related to both aging and metabolic disease. Calorie restriction (CR) can retard the aging process in organisms ranging from yeast to rodents and delay the onset of numerous age-related disorders, such as insulin resistance and diabetes. Therefore, metabolic CR mimetics may represent new therapeutic targets for insulin resistance and T2DM. Sirtuin 1 (SIRT1), the mammalian homolog of Sir2, was originally identified as a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase. The activation of SIRT1 is closely associated with longevity under CR, and it is recognized as a CR mimetic. Currently, seven sirtuins have been identified in mammals. Among these sirtuins, SIRT1 and SIRT2 are located in the nucleus and cytoplasm, SIRT3 exists predominantly in mitochondria, and SIRT6 is located in the nucleus. These sirtuins regulate metabolism through their regulation of inflammation, oxidative stress and mitochondrial function via multiple mechanisms, resulting in the improvement of insulin resistance and T2DM. In this review, we describe the current understanding of the biological functions of sirtuins, especially SIRT1, SIRT2, SIRT3, and SIRT6, focusing on oxidative stress, inflammation, and mitochondrial function, which are closely associated with aging.
Collapse
Affiliation(s)
- Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
- *Correspondence: Munehiro Kitada
| | - Yoshio Ogura
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Itaru Monno
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
99
|
Bruns I, Sauer B, Burger MC, Eriksson J, Hofmann U, Braun Y, Harter PN, Luger AL, Ronellenfitsch MW, Steinbach JP, Rieger J. Disruption of peroxisome proliferator-activated receptor γ coactivator (PGC)-1α reverts key features of the neoplastic phenotype of glioma cells. J Biol Chem 2018; 294:3037-3050. [PMID: 30578297 DOI: 10.1074/jbc.ra118.006993] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Indexed: 12/30/2022] Open
Abstract
The peroxisome proliferator-activated receptor γ coactivator (PGC)-1α is a master regulator of mitochondrial biogenesis and controls metabolism by coordinating transcriptional events. Here, we interrogated whether PGC-1α is involved in tumor growth and the metabolic flexibility of glioblastoma cells. PGC-1α was expressed in a subset of established glioma cell lines and primary glioblastoma cell cultures. Furthermore, a higher PGC-1α expression was associated with an adverse outcome in the TCGA glioblastoma dataset. Suppression of PGC-1α expression by shRNA in the PGC-1α-positive U343MG glioblastoma line suppressed mitochondrial gene expression, reduced mitochondrial membrane potential, and diminished oxygen as well as glucose consumption, and lactate production. Compatible with the known PGC-1α functions in reactive oxygen species (ROS) metabolism, glioblastoma cells deficient in PGC-1α displayed ROS accumulation, had reduced RNA levels of proteins involved in ROS detoxification, and were more susceptible to death induction by H2O2 compared with control cells. PGC-1αsh cells also had impaired proliferation and migration rates in vitro and displayed less stem cell characteristics. Complementary effects were observed in PGC-1α-low LNT-229 cells engineered to overexpress PGC-1α. In an in vivo xenograft experiment, tumors formed by U343MG PGC-1αsh glioblastoma cells grew much slower than control tumors and were less invasive. Interestingly, the PGC-1α knockdown conferred protection against hypoxia-induced cell death, probably as a result of less active anabolic pathways, and this effect was associated with reduced epidermal growth factor expression and mammalian target of rapamycin signaling. In summary, PGC-1α modifies the neoplastic phenotype of glioblastoma cells toward more aggressive behavior and therefore makes PGC-1α a potential target for anti-glioblastoma therapies.
Collapse
Affiliation(s)
- Ines Bruns
- From the Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany.,the German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt.,the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,the University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Benedikt Sauer
- From the Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany.,the German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt.,the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,the University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Michael C Burger
- From the Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany.,the German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt.,the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,the University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Jule Eriksson
- From the Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany.,the Department of Neurology, University of Basel, 4031 Basel, Switzerland
| | - Ute Hofmann
- the Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany.,the University of Tübingen, 72074 Tübingen, Germany
| | - Yannick Braun
- the Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany, and
| | - Patrick N Harter
- the German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt.,the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,the University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany.,the Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany, and
| | - Anna-Luisa Luger
- From the Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany.,the German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt.,the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,the University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Michael W Ronellenfitsch
- From the Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany, .,the German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt.,the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,the University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Joachim P Steinbach
- From the Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany, .,the German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt.,the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,the University Cancer Center (UCT), University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Johannes Rieger
- From the Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany.,the Interdisciplinary Division of Neuro-Oncology, Hertie Institute of Clinical Brain Research, University Hospital Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
100
|
Blanco CL, Gastaldelli A, Anzueto DG, Winter LA, Seidner SR, McCurnin DC, Liang H, Javors MA, DeFronzo RA, Musi N. Effects of intravenous AICAR (5-aminoimidazole-4-carboximide riboside) administration on insulin signaling and resistance in premature baboons, Papio sp. PLoS One 2018; 13:e0208757. [PMID: 30540820 PMCID: PMC6291136 DOI: 10.1371/journal.pone.0208757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022] Open
Abstract
Premature baboons exhibit peripheral insulin resistance and impaired insulin signaling. 5' AMP-activated protein kinase (AMPK) activation improves insulin sensitivity by enhancing glucose uptake (via increased glucose transporter type 4 [GLUT4] translocation and activation of the extracellular signal-regulated kinase [ERK]/ atypical protein kinase C [aPKC] pathway), and increasing fatty acid oxidation (via inhibition of acetyl-CoA carboxylase 1 [ACC]), while downregulating gluconeogenesis (via induction of small heterodimer partner [SHP] and subsequent downregulation of the gluconeogenic enzymes: phosphoenolpyruvate carboxykinase [PEPCK], glucose 6-phosphatase [G6PASE], fructose- 1,6-bisphosphatase 1 [FBP1], and forkhead box protein 1 [FOXO1]). The purpose of this study was to investigate whether pharmacologic activation of AMPK with AICAR (5-aminoimidazole-4-carboximide riboside) administration improves peripheral insulin sensitivity in preterm baboons. 11 baboons were delivered prematurely at 125±2 days (67%) gestation. 5 animals were randomized to receive 5 days of continuous AICAR infusion at a dose of 0.5 mg·g-1·day-1. 6 animals were in the placebo group. Euglycemic hyperinsulinemic clamps were performed at 5±2 and 14±2 days of life. Key molecules potentially altered by AICAR (AMPK, GLUT4, ACC, PEPCK, G6PASE, FBP1, and FOXO1), and the insulin signaling molecules: insulin receptor (INSR), insulin receptor substrate 1 (IRS-1), protein kinase B (AKT), and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) were measured using RT-PCR and western blotting. AICAR infusion did not improve whole body insulin-stimulated glucose disposal in preterm baboons (12.8±2.4 vs 12.4±2.0 mg/(kg·min), p = 0.8, placebo vs AICAR). One animal developed complications during treatment. In skeletal muscle, AICAR infusion did not increase phosphorylation of ACC, AKT, or AMPK whereas it increased mRNA expression of ACACA (ACC), AKT, and PPARGC1A (PGC1α). In the liver, INSR, IRS1, G6PC3, AKT, PCK1, FOXO1, and FBP1 were unchanged, whereas PPARGC1A mRNA expression increased after AICAR infusion. This study provides evidence that AICAR does not improve insulin sensitivity in premature euglycemic baboons, and may have adverse effects.
Collapse
Affiliation(s)
- Cynthia L. Blanco
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Amalia Gastaldelli
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Institute of Clinical Physiology Consiglio Nazionale delle Ricerche, Pisa Italy
| | - Diana G. Anzueto
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Lauryn A. Winter
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Steven R. Seidner
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Donald C. McCurnin
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Hanyu Liang
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Martin A. Javors
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Ralph A. DeFronzo
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Texas Diabetes Institute, San Antonio, TX, United States of America
| | - Nicolas Musi
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Texas Diabetes Institute, San Antonio, TX, United States of America
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, United States of America
| |
Collapse
|