1
|
Ji X, Zhang C, Yang J, Tian Y, You L, Yang H, Li Y, Liu H, Pan D, Liu Z. Kaempferol Improves Exercise Performance by Regulating Glucose Uptake, Mitochondrial Biogenesis, and Protein Synthesis via PI3K/AKT and MAPK Signaling Pathways. Foods 2024; 13:1068. [PMID: 38611372 PMCID: PMC11011654 DOI: 10.3390/foods13071068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Kaempferol is a natural flavonoid with reported bioactivities found in many fruits, vegetables, and medicinal herbs. However, its effects on exercise performance and muscle metabolism remain inconclusive. The present study investigated kaempferol's effects on improving exercise performance and potential mechanisms in vivo and in vitro. The grip strength, exhaustive running time, and distance of mice were increased in the high-dose kaempferol group (p < 0.01). Also, kaempferol reduced fatigue-related biochemical markers and increased the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) related to antioxidant capacity. Kaempferol also increased the glycogen and adenosine triphosphate (ATP) content in the liver and skeletal muscle, as well as glucose in the blood. In vitro, kaempferol promoted glucose uptake, protein synthesis, and mitochondrial function and decreased oxidative stress in both 2D and 3D C2C12 myotube cultures. Moreover, kaempferol activated the PI3K/AKT and MAPK signaling pathways in the C2C12 cells. It also upregulated the key targets of glucose uptake, mitochondrial function, and protein synthesis. These findings suggest that kaempferol improves exercise performance and alleviates physical fatigue by increasing glucose uptake, mitochondrial biogenesis, and protein synthesis and by decreasing ROS. Kaempferol's molecular mechanism may be related to the regulation of the PI3K/AKT and MAPK signaling pathways.
Collapse
Affiliation(s)
- Xiaoning Ji
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Chaozheng Zhang
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Jing Yang
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Yaru Tian
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Lijuan You
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Hui Yang
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Yongning Li
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Haibo Liu
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Deng Pan
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| | - Zhaoping Liu
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China (H.Y.); (Z.L.)
| |
Collapse
|
2
|
Ji X, Li Q, Liu Z, Wu W, Zhang C, Sui H, Chen M. Identification of Active Components for Sports Supplements: Machine Learning-Driven Classification and Cell-Based Validation. ACS OMEGA 2024; 9:11347-11355. [PMID: 38496927 PMCID: PMC10938306 DOI: 10.1021/acsomega.3c07395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 03/19/2024]
Abstract
The identification of active components is critical for the development of sports supplements. However, high-throughput screening of active components remains a challenge. This study sought to construct prediction models to screen active components from herbal medicines via machine learning and validate the screening by using cell-based assays. The six constructed models had an accuracy of >0.88. Twelve randomly selected active components from the screening were tested for their active potency on C2C12 cells, and 11 components induced a significant increase in myotube diameters and protein synthesis. The effect and mechanism of luteolin among the 11 active components as potential sports supplements were then investigated by using immunofluorescence staining and high-content imaging analysis. It showed that luteolin increased the skeletal muscle performance via the activation of PGC-1α and MAPK signaling pathways. Thus, high-throughput prediction models can be effectively used to screen active components as sports supplements.
Collapse
Affiliation(s)
- Xiaoning Ji
- State
Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di
Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- NHC
key laboratory of food safety risk assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Qiuyun Li
- NMPA
Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial
Key Laboratory of Tropical Disease Research, Food Safety and Health
Research Center, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Zhaoping Liu
- NHC
key laboratory of food safety risk assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Weiliang Wu
- NMPA
Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial
Key Laboratory of Tropical Disease Research, Food Safety and Health
Research Center, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Chaozheng Zhang
- NHC
key laboratory of food safety risk assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Haixia Sui
- NHC
key laboratory of food safety risk assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Min Chen
- State
Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di
Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
3
|
Li F, Xu M, Miao J, Hu N, Wang Y, Wang L. Down-regulated Smyd1 participated in the inhibition of myoblast differentiation induced by cigarette smoke extract. Toxicol Lett 2023; 383:S0378-4274(23)00211-4. [PMID: 37385529 DOI: 10.1016/j.toxlet.2023.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 05/25/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
The histone methyltransferase Smyd1 is essential for muscle development; however, its role in smoking-induced skeletal muscle atrophy and dysfunction has not been investigated thus far. In this study, Smyd1 was overexpressed or knocked down in C2C12 myoblasts by an adenovirus vector and cultured in differentiation medium containing 5% cigarette smoke extract (CSE) for 4 days. CSE exposure resulted in inhibition of C2C12 cell differentiation and downregulation of Smyd1 expression, whereas Smyd1 overexpression reduced the degree of inhibition of myotube differentiation caused by CSE exposure. CSE exposure activated P2RX7-mediated apoptosis and pyroptosis, caused increased intracellular reactive oxygen species (ROS) levels, and impaired mitochondrial biogenesis and increased protein degradation by downregulating PGC1α, whereas Smyd1 overexpression partially restored the altered protein levels caused by CSE exposure. Smyd1 knockdown alone produced a phenotype similar to CSE exposure, and Smyd1 knockdown during CSE exposure aggravated the degree of inhibition of myotube differentiation and the degree of activation of P2RX7. CSE exposure suppressed H3K4me2 expression, and chromatin immunoprecipitation confirmed the transcriptional regulation of P2rx7 by H3K4me2 modification. Our findings suggest that CSE exposure mediates C2C12 cell apoptosis and pyroptosis through the Smyd1-H3K4me2-P2RX7 axis, and inhibits PGC1α expression to impair mitochondrial biosynthesis and increase protein degradation by inhibiting Smyd1 expression, ultimately leading to abnormal C2C12 myoblasts differentiation and impaired myotube formation.
Collapse
Affiliation(s)
- Fang Li
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province
| | - Mengting Xu
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province
| | - Jianing Miao
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province
| | - Nengyin Hu
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province
| | - Ying Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| | - Lili Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China; Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, 110004, China; Key Laboratory of Research and Application of Animal Model for Environmental and Metabolic Diseases, Liaoning Province.
| |
Collapse
|
4
|
Kang Z, Wu B, Zhang L, Liang X, Guo D, Yuan S, Xie D. Metabolic regulation by biomaterials in osteoblast. Front Bioeng Biotechnol 2023; 11:1184463. [PMID: 37324445 PMCID: PMC10265685 DOI: 10.3389/fbioe.2023.1184463] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023] Open
Abstract
The repair of bone defects resulting from high-energy trauma, infection, or pathological fracture remains a challenge in the field of medicine. The development of biomaterials involved in the metabolic regulation provides a promising solution to this problem and has emerged as a prominent research area in regenerative engineering. While recent research on cell metabolism has advanced our knowledge of metabolic regulation in bone regeneration, the extent to which materials affect intracellular metabolic remains unclear. This review provides a detailed discussion of the mechanisms of bone regeneration, an overview of metabolic regulation in bone regeneration in osteoblasts and biomaterials involved in the metabolic regulation for bone regeneration. Furthermore, it introduces how materials, such as promoting favorable physicochemical characteristics (e.g., bioactivity, appropriate porosity, and superior mechanical properties), incorporating external stimuli (e.g., photothermal, electrical, and magnetic stimulation), and delivering metabolic regulators (e.g., metal ions, bioactive molecules like drugs and peptides, and regulatory metabolites such as alpha ketoglutarate), can affect cell metabolism and lead to changes of cell state. Considering the growing interests in cell metabolic regulation, advanced materials have the potential to help a larger population in overcoming bone defects.
Collapse
Affiliation(s)
- Zhengyang Kang
- Department of Orthopedics, The Second People’s Hospital of Panyu Guangzhou, Guangzhou, China
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Bin Wu
- Department of Orthopedics, The Second People’s Hospital of Panyu Guangzhou, Guangzhou, China
| | - Luhui Zhang
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xinzhi Liang
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dong Guo
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shuai Yuan
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Denghui Xie
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangxi Key Laboratory of Bone and Joint Degeneration Diseases, Youjiang Medical University For Nationalities, Baise, China
| |
Collapse
|
5
|
Li Y, Zhang S, Huang C, Lin D. NMR-Based Metabolic Profiling of the Effects of α-Ketoglutarate Supplementation on Energy-Deficient C2C12 Myotubes. Molecules 2023; 28:molecules28093840. [PMID: 37175250 PMCID: PMC10179873 DOI: 10.3390/molecules28093840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Skeletal muscle is closely linked to energy metabolism, but it is inevitably deprived of energy. Cellular differentiation is an essential and energy-demanding process in skeletal muscle development. Much attention has been paid to identifying beneficial factors that promote skeletal muscle satellite cell differentiation and further understanding the underlying regulatory mechanisms. As a critical metabolic substrate or regulator, α-ketoglutarate (AKG) has been recognized as a potential nutritional supplement or therapeutic target for skeletal muscle. We have previously found beneficial effects of AKG supplementation on the proliferation of C2C12 myoblasts cultured under both normal and energy-deficient conditions and have further elucidated the underlying metabolic mechanisms. However, it remains unclear what role AKG plays in myotube formation in different energy states. In the present study, we investigated the effects of AKG supplementation on the differentiation of C2C12 myoblasts cultured in normal medium (Nor myotubes) and low glucose medium (Low myotubes) and performed NMR-based metabonomic profiling to address AKG-induced metabolic changes in both Nor and Low myotubes. Significantly, AKG supplementation promoted myotube formation and induced metabolic remodeling in myotubes under normal medium and low glucose medium, including improved energy metabolism and enhanced antioxidant capacity. Specifically, AKG mainly altered amino acid metabolism and antioxidant metabolism and upregulated glycine levels and antioxidase expression. Our results are typical for the mechanistic understanding of the effects of AKG supplementation on myotube formation in the two energy states. This study may be beneficial for further exploring the applications of AKG supplementation in sports, exercise, and therapy.
Collapse
Affiliation(s)
- Yantong Li
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuya Zhang
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361021, China
| | - Donghai Lin
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
6
|
Lee H, Kim SY, Lim Y. Lespedeza bicolor extract supplementation reduced hyperglycemia-induced skeletal muscle damage by regulation of AMPK/SIRT/PGC1α-related energy metabolism in type 2 diabetic mice. Nutr Res 2023; 110:1-13. [PMID: 36638746 DOI: 10.1016/j.nutres.2022.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Lespedeza bicolor (LB) is known to have antidiabetic activities; however, the underlying molecular mechanisms of LB in hyperglycemia-induced skeletal muscle damage is unclear. Inflammation and oxidative stress caused by type 2 diabetes mellitus (T2DM) not only contributes to insulin resistance, but also promotes muscle atrophy via decreased muscle protein synthesis and increased protein degradation, leading to frailty and sarcopenia. In this study, we hypothesized that LB extract (LBE) supplementatin has an ameliorative effect on hyperglycemia-induced skeletal muscle damage by activation of 5' adenosine monophosphate-activated protein kinase (AMPK)/sirtuin (SIRT)/proliferator-activated receptor γ coactivator 1α (PGC1α)-associated energy metabolism in mice with T2DM. Diabetes was induced by a high-fat diet with a 2-time streptozotoxin injection (30 mg/kg body weight) in male C57BL/6J mice. After diabetes was induced (fasting blood glucose level ≥140 mg/dL), the mice were administered with LBE at a low dose (100 mg/kg/d) or high dose (250 mg/kg/d) by gavage for 12 weeks. LBE supplementation ameliorated glucose tolerance and hemoglobin A1c (%) in mice with T2DM. Moreover, LBE supplementation upregulated protein levels of insulin receptor subunit-1 and Akt accompanied by increased translocation of glucose transporter 4 in mice with T2DM. Furthermore, LBE increased mitochondrial biogenesis by activating SIRT1, SIRT3, SIRT4, and peroxisome PGC1α in diabetic skeletal muscle. Meanwhile, LBE supplementation reduced oxidative stress and inflammation in mice with T2DM. Taken together, the current study suggested that LBE could be a potential therapeutic to prevent skeletal muscle damage by regulation AMPK/SIRT/PGC1α-related energy metabolism in T2DM.
Collapse
Affiliation(s)
- Heaji Lee
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Yunsook Lim
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
7
|
Dai W, Liu K, Li R, Cao Y, Shen M, Tao J, Liu H. Trillin inhibits myoblast differentiation via increasing autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153962. [PMID: 35172256 DOI: 10.1016/j.phymed.2022.153962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/07/2022] [Accepted: 01/25/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Trillin, an active ingredient in traditional Chinese medicine Trillium tschonoskii, is a potential small molecule compound candidate that affecting myoblast differentiation, which predicting by AI technology in our previous study. Autophagy modulating myoblast differentiation has also been studied. In addition, Trillin was shown to regulate mTOR signaling pathway, a highly conserved kinase important for autophagy regulation. PURPOSE In this research, we aim to clarify the effect and underlying mechanism of Trillin on myoblast differentiation. STUDY DESIGN AND METHODS Using mice C2C12 cell line to establish a myoblast differentiation model in vitro, treated with different concentration and time of Trillin, to explore the effect and latent mechanism of Trillin on myoblast differentiation by qRT-PCR, Western Blot and other molecular biological technique. RESULTS Results showed that C2C12 differentiation was significantly inhibited by Trillin in a dose-dependent manner. The expression of MyHC, MyOG and MyoD was decreased extremely significant after 10 μM Trillin treatment. Meanwhile, autophagy level was significantly elevated with the supplement of Trillin. And C2C12 differentiation was recovered after ATG7 knockdown. Mechanically, we found that the activity of AKT/mTOR declined during the inhibition of differentiation by Trillin. CONCLUSION Our findings suggested that Trillin attenuated C2C12 differentiation via increasing autophagy through AKT/mTOR signaling pathway. Taken together, we introduce a novel physiological function of Trillin in inhibiting skeletal muscle differentiation.
Collapse
Affiliation(s)
- Weilong Dai
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ke Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Rongyang Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yan Cao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ming Shen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jingli Tao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Honglin Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
8
|
Liang S, Liu X, Zhao J, Liu R, Huang X, Liu Y, Yang X, Yang X. Effects of high-dose folic acid on protein metabolism in breast muscle and performance of broilers. Poult Sci 2022; 101:101935. [PMID: 35961252 PMCID: PMC9382563 DOI: 10.1016/j.psj.2022.101935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
Attaining the optimal feed conversion ratio is the unaltered goal for poultry breeding, meat yield is one of the vital reference indexes for that. Folic acid is involved in protein metabolism by acting as a transmitter of one carbon unit, and the detail mechanism for the high-dose folic acid on growth of broiler skeletal muscle is still unclarified. The present study was conducted to investigate the effect and regulatory mechanism of folic acid on deposition and metabolism of protein in broiler breast muscle. A total of 196 one-day-old AA broilers were randomly assigned to 2 treatment groups. The chicks were fed corn-soybean diet with folic acid levels of 1.3 mg/kg (CON) or 13 mg/kg (FA), respectively. The results showed that high dose of folic acid significantly increased the body weight gain, average daily gain, average daily feed intake, and feed conversion ratio of broilers during 1 to 42 d. Compared with control group, folic acid statistically augmented the breast muscle ratio of broilers at 42 d, abdominal fat percentage was also decreased in FA group. Folic acid significantly increased the gene expression of folate receptor (FR) in duodenum and jejunum at 21 d, and its relative expression in jejunum of broilers at 42 d. Furthermore, relative expression of myogenin in broiler breast muscle was upregulated in folic acid group. Folic acid supplementation significantly enhanced the protein expression of phosphorylated serine/threonine kinase (AKT) and ribosomal protein S6 kinase 1 (S6K1) in the breast muscle of broilers at 21 d and 42 d. In conclusion, the results proved that high-dose folic acid activated the AKT/mammalian target of rapamycin (mTOR) pathway and increased the activity of phosphorylation of S6K1, thereby regulating the protein deposition in breast muscle. Meanwhile, the gene expression of the myogenic determinant factor was upregulated by folic acid and then promoted the growth of breast muscle. Consequently, the growth performance, meat production and feeding efficiency were improved of broilers by adding folic acid at 13 mg/kg.
Collapse
Affiliation(s)
- Saisai Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinshuai Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jianfei Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rui Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinhuo Huang
- Nano Vitamin Engineering Research Center of Shaanxi Province, Xi'an, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.
| |
Collapse
|
9
|
MORISASA M, YOSHIDA E, FUJITANI M, KIMURA K, UCHIDA K, KISHIDA T, MORI T, GOTO-INOUE N. Fish Protein Promotes Skeletal Muscle Hypertrophy via the Akt/mTOR Signaling Pathways. J Nutr Sci Vitaminol (Tokyo) 2022; 68:23-31. [DOI: 10.3177/jnsv.68.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Mizuki MORISASA
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University
| | - Eriko YOSHIDA
- Functional Ingredient Research Section, Food Function R&D Center, Nippon Suisan Kaisha, Ltd
| | - Mina FUJITANI
- Laboratory of Nutrition Science, Division of Applied Bioscience, Graduate School of Agriculture, Ehime University
| | - Keisuke KIMURA
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University
| | - Kenji UCHIDA
- Functional Ingredient Research Section, Food Function R&D Center, Nippon Suisan Kaisha, Ltd
| | - Taro KISHIDA
- Laboratory of Nutrition Science, Division of Applied Bioscience, Graduate School of Agriculture, Ehime University
| | - Tsukasa MORI
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University
| | - Naoko GOTO-INOUE
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University
| |
Collapse
|
10
|
Nanohydroxyapatite-Blasted Bioactive Surface Drives Shear-Stressed Endothelial Cell Growth and Angiogenesis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1433221. [PMID: 35252440 PMCID: PMC8890866 DOI: 10.1155/2022/1433221] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/23/2021] [Accepted: 01/19/2022] [Indexed: 12/22/2022]
Abstract
Nanosized crystalline hydroxyapatite coating (HAnano®) accelerates the osteointegration of dental implants which is hypothesized to drive angiogenesis. In order to test this hypothesis, we have subjected shear-stressed human umbilical vein endothelial cells (HUVECs) to a HAnano®-enriched medium, as well as to surface presenting dual acid etching (DAE) as a control. To note, the titanium implants were coated with 10 nm in diameter HA particles using the Promimic HAnano method. Our data reveals that HAnano® modulates higher expression of genes related with endothelial cell performance and viability, such as VEGF, eNOS, and AKT, and further angiogenesis in vitro by promoting endothelial cell migration. Additionally, the data shows a significant extracellular matrix (ECM) remodeling, and this finding seems developing a dual role in promoting the expression of VEGF and control endothelial cell growth during angiogenesis. Altogether, these data prompted us to further validate this phenomenon by exploring genes related with the control of cell cycle and in fact our data shows that HAnano® promotes higher expression of CDK4 gene, while p21 and p15 genes (suppressor genes) were significantly lower. In conjunction, our data shows for the first time that HAnano®-coated surfaces drive angiogenesis by stimulating a proliferative and migration phenotype of endothelial cells, and this finding opens novel comprehension about osseointegration mechanism considering nanosized hydroxyapatite coating dental implants.
Collapse
|
11
|
Hayashi T, Kudo T, Fujita R, Fujita SI, Tsubouchi H, Fuseya S, Suzuki R, Hamada M, Okada R, Muratani M, Shiba D, Suzuki T, Warabi E, Yamamoto M, Takahashi S. Nuclear factor E2-related factor 2 (NRF2) deficiency accelerates fast fibre type transition in soleus muscle during space flight. Commun Biol 2021; 4:787. [PMID: 34168270 PMCID: PMC8225765 DOI: 10.1038/s42003-021-02334-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/20/2021] [Indexed: 12/20/2022] Open
Abstract
Microgravity induces skeletal muscle atrophy, particularly in the soleus muscle, which is predominantly composed of slow-twitch myofibre (type I) and is sensitive to disuse. Muscle atrophy is commonly known to be associated with increased production of reactive oxygen species. However, the role of NRF2, a master regulator of antioxidative response, in skeletal muscle plasticity during microgravity-induced atrophy, is not known. To investigate the role of NRF2 in skeletal muscle within a microgravity environment, wild-type and Nrf2-knockout (KO) mice were housed in the International Space Station for 31 days. Gene expression and histological analyses demonstrated that, under microgravity conditions, the transition of type I (oxidative) muscle fibres to type IIa (glycolytic) was accelerated in Nrf2-KO mice without affecting skeletal muscle mass. Therefore, our results suggest that NRF2 affects myofibre type transition during space flight.
Collapse
Affiliation(s)
- Takuto Hayashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Takashi Kudo
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.
| | - Ryo Fujita
- Divsion of Regenerative Medicine, Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Shin-Ichiro Fujita
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan.,Department of Genome Biology, Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hirona Tsubouchi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Sayaka Fuseya
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Riku Suzuki
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki, Japan
| | - Michito Hamada
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Risa Okada
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Ibaraki, Japan
| | - Masafumi Muratani
- Department of Genome Biology, Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Dai Shiba
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Ibaraki, Japan
| | - Takafumi Suzuki
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Eiji Warabi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, and Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.
| |
Collapse
|
12
|
Li Y, Li X, Gao Y, Huang C, Lin D. NMR-Based Metabolomic Analysis for the Effects of α-Ketoglutarate Supplementation on C2C12 Myoblasts in Different Energy States. Molecules 2021; 26:1841. [PMID: 33805924 PMCID: PMC8037044 DOI: 10.3390/molecules26071841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 11/17/2022] Open
Abstract
α-Ketoglutarate (AKG) is attracting much attention from researchers owing to its beneficial effects on anti-aging and cancer suppression, and, more recently, in nutritional supplements. Given that glucose is the main source of energy to maintain normal physiological functions of skeletal muscle, the effects of AKG supplementation for improving muscle performance are closely related to the glucose level in skeletal muscle. The differences of AKG-induced effects in skeletal muscle between two states of normal energy and energy deficiency are unclear. Furthermore, AKG-induced metabolic changes in skeletal muscles in different energy states also remain elusive. Here, we assessed the effects of AKG supplementation on mouse C2C12 myoblast cells cultured both in normal medium (Nor cells) and in low-glucose medium (Low cells), which were used to mimic two states of normal energy and energy deficiency, respectively. We further performed NMR-based metabolomic analysis to address AKG-induced metabolic changes in Nor and Low cells. AKG supplementation significantly promoted the proliferation and differentiation of cells in the two energy states through glutamine metabolism, oxidative stress, and energy metabolism. Under normal culture conditions, AKG up-regulated the intracellular glutamine level, changed the cellular energy status, and maintained the antioxidant capacity of cells. Under low-glucose culture condition, AKG served as a metabolic substrate to reduce the glutamine-dependence of cells, remarkably enhanced the antioxidant capacity of cells and significantly elevated the intracellular ATP level, thereby ensuring the normal growth and metabolism of cells in the state of energy deficiency. Our results provide a mechanistic understanding of the effects of AKG supplements on myoblasts in both normal energy and energy deficiency states. This work may be beneficial to the exploitation of AKG applications in clinical treatments and nutritional supplementations.
Collapse
Affiliation(s)
- Yantong Li
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Xiaoyuan Li
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Yifeng Gao
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361021, China
| | - Donghai Lin
- Key Laboratory of Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.L.); (X.L.); (Y.G.)
| |
Collapse
|
13
|
Pleiotropic effects of alpha-ketoglutarate as a potential anti-ageing agent. Ageing Res Rev 2021; 66:101237. [PMID: 33340716 DOI: 10.1016/j.arr.2020.101237] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/23/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
An intermediate of tricarboxylic acid cycle alpha-ketoglutarate (AKG) is involved in pleiotropic metabolic and regulatory pathways in the cell, including energy production, biosynthesis of certain amino acids, collagen biosynthesis, epigenetic regulation of gene expression, regulation of redox homeostasis, and detoxification of hazardous substances. Recently, AKG supplement was found to extend lifespan and delay the onset of age-associated decline in experimental models such as nematodes, fruit flies, yeasts, and mice. This review summarizes current knowledge on metabolic and regulatory functions of AKG and its potential anti-ageing effects. Impact on epigenetic regulation of ageing via being an obligate substrate of DNA and histone demethylases, direct antioxidant properties, and function as mimetic of caloric restriction and hormesis-induced agent are among proposed mechanisms of AKG geroprotective action. Due to influence on mitochondrial respiration, AKG can stimulate production of reactive oxygen species (ROS) by mitochondria. According to hormesis hypothesis, moderate stimulation of ROS production could have rather beneficial biological effects, than detrimental ones, because of the induction of defensive mechanisms that improve resistance to stressors and age-related diseases and slow down functional senescence. Discrepancies found in different models and limitations of AKG as a geroprotective drug are discussed.
Collapse
|
14
|
Shen J, Hao Z, Wang J, Hu J, Liu X, Li S, Ke N, Song Y, Lu Y, Hu L, Qiao L, Wu X, Luo Y. Comparative Transcriptome Profile Analysis of Longissimus dorsi Muscle Tissues From Two Goat Breeds With Different Meat Production Performance Using RNA-Seq. Front Genet 2021; 11:619399. [PMID: 33519920 PMCID: PMC7838615 DOI: 10.3389/fgene.2020.619399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Carcass weight, meat quality and muscle components are important traits economically and they underpin most of the commercial return to goat producers. In this study, the Longissimus dorsi muscle tissues were collected from five Liaoning cashmere (LC) goats and five Ziwuling black (ZB) goats with phenotypic difference in carcass weight, some meat quality traits and muscle components. The histological quantitative of collagen fibers and the transcriptome profiles in the Longissimus dorsi muscle tissues were investigated using Masson-trichrome staining and RNA-Seq, respectively. The percentage of total collagen fibers in the Longissimus dorsi muscle tissues from ZB goats was less than those from LC goats, suggesting that these ZB goats had more tender meat. An average of 15,919 and 15,582 genes were found to be expressed in Longissimus dorsi muscle tissues from LC and ZB goats, respectively. Compared to LC goats, the expression levels of 78 genes were up-regulated in ZB goats, while 133 genes were down-regulated. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that the differentially expressed genes (DEGs) were significantly enriched in GO terms related to the muscle growth and development and the deposition of intramuscular fat and lipid metabolism, hippo signaling pathway and Jak-STAT signaling pathway. The results provide an improved understanding of the genetic mechanisms regulating meat production performance in goats, and will help us improve the accuracy of selection for meat traits in goats using marker-assisted selection based on these differentially expressed genes obtained.
Collapse
Affiliation(s)
- Jiyuan Shen
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Zhiyun Hao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jiqing Wang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiu Liu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shaobin Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Na Ke
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yize Song
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yujie Lu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Liyan Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Lirong Qiao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xinmiao Wu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yuzhu Luo
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
15
|
Sartori R, Romanello V, Sandri M. Mechanisms of muscle atrophy and hypertrophy: implications in health and disease. Nat Commun 2021; 12:330. [PMID: 33436614 PMCID: PMC7803748 DOI: 10.1038/s41467-020-20123-1] [Citation(s) in RCA: 491] [Impact Index Per Article: 122.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/16/2020] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle is the protein reservoir of our body and an important regulator of glucose and lipid homeostasis. Consequently, the growth or the loss of muscle mass can influence general metabolism, locomotion, eating and respiration. Therefore, it is not surprising that excessive muscle loss is a bad prognostic index of a variety of diseases ranging from cancer, organ failure, infections and unhealthy ageing. Muscle function is influenced by different quality systems that regulate the function of contractile proteins and organelles. These systems are controlled by transcriptional dependent programs that adapt muscle cells to environmental and nutritional clues. Mechanical, oxidative, nutritional and energy stresses, as well as growth factors or cytokines modulate signaling pathways that, ultimately, converge on protein and organelle turnover. Novel insights that control and orchestrate such complex network are continuously emerging and will be summarized in this review. Understanding the mechanisms that control muscle mass will provide therapeutic targets for the treatment of muscle loss in inherited and non-hereditary diseases and for the improvement of the quality of life during ageing. Loss of muscle mass is associated with ageing and with a number of diseases such as cancer. Here, the authors review the signaling pathways that modulate protein synthesis and degradation and gain or loss of muscle mass, and discuss therapeutic implications and future directions for the field.
Collapse
Affiliation(s)
- Roberta Sartori
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35121, Padova, Italy.,Veneto Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy
| | - Vanina Romanello
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35121, Padova, Italy. .,Veneto Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy.
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35121, Padova, Italy. .,Veneto Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy. .,Myology Center, University of Padova, via Ugo Bassi 58/b, 35121, Padova, Italy. .,Department of Medicine, McGill University, Montreal, Canada.
| |
Collapse
|
16
|
Yang YR, Kwon KS. Potential Roles of Exercise-Induced Plasma Metabolites Linking Exercise to Health Benefits. Front Physiol 2020; 11:602748. [PMID: 33343398 PMCID: PMC7744613 DOI: 10.3389/fphys.2020.602748] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/19/2020] [Indexed: 12/30/2022] Open
Abstract
Regular exercise has a myriad of health benefits. An increase in circulating exercise factors following exercise is a critical physiological response. Numerous studies have shown that exercise factors released from tissues during physical activity may contribute to health benefits via autocrine, paracrine, and endocrine mechanisms. Myokines, classified as proteins secreted from skeletal muscle, are representative exercise factors. The roles of myokines have been demonstrated in a variety of exercise-related functions linked to health benefits. In addition to myokines, metabolites are also exercise factors. Exercise changes the levels of various metabolites via metabolic reactions. Several studies have identified exercise-induced metabolites that positively influence organ functions. Here, we provide an overview of selected metabolites secreted into the circulation upon exercise.
Collapse
Affiliation(s)
- Yong Ryoul Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| |
Collapse
|
17
|
Yang W, Huang J, Wu H, Wang Y, Du Z, Ling Y, Wang W, Wu Q, Gao W. Molecular mechanisms of cancer cachexia‑induced muscle atrophy (Review). Mol Med Rep 2020; 22:4967-4980. [PMID: 33174001 PMCID: PMC7646947 DOI: 10.3892/mmr.2020.11608] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022] Open
Abstract
Muscle atrophy is a severe clinical problem involving the loss of muscle mass and strength that frequently accompanies the development of numerous types of cancer, including pancreatic, lung and gastric cancers. Cancer cachexia is a multifactorial syndrome characterized by a continuous decline in skeletal muscle mass that cannot be reversed by conventional nutritional therapy. The pathophysiological characteristic of cancer cachexia is a negative protein and energy balance caused by a combination of factors, including reduced food intake and metabolic abnormalities. Numerous necessary cellular processes are disrupted by the presence of abnormal metabolites, which mediate several intracellular signaling pathways and result in the net loss of cytoplasm and organelles in atrophic skeletal muscle during various states of cancer cachexia. Currently, the clinical morbidity and mortality rates of patients with cancer cachexia are high. Once a patient enters the cachexia phase, the consequences are difficult to reverse and the treatment methods for cancer cachexia are very limited. The present review aimed to summarize the recent discoveries regarding the pathogenesis of cancer cachexia-induced muscle atrophy and provided novel ideas for the comprehensive treatment to improve the prognosis of affected patients.
Collapse
Affiliation(s)
- Wei Yang
- Department of Oncology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Jianhui Huang
- Department of Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Hui Wu
- Department of Clinical Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yuqing Wang
- Department of Clinical Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Zhiyin Du
- Department of Clinical Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yuanbo Ling
- Department of Clinical Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Weizhuo Wang
- Department of Clinical Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Qian Wu
- Department of Oncology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Wenbin Gao
- Department of Oncology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
18
|
Chen W, Wang L, You W, Shan T. Myokines mediate the cross talk between skeletal muscle and other organs. J Cell Physiol 2020; 236:2393-2412. [PMID: 32885426 DOI: 10.1002/jcp.30033] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Myokines are muscle-derived cytokines and chemokines that act extensively on organs and exert beneficial metabolic functions in the whole-body through specific signal networks. Myokines as mediators provide the conceptual basis for a whole new paradigm useful for understanding how skeletal muscle communicates with other organs. In this review, we summarize and discuss classes of myokines and their physiological functions in mediating the regulatory roles of skeletal muscle on other organs and the regulation of the whole-body energy metabolism. We review the mechanisms involved in the interaction between skeletal muscle and nonmuscle organs through myokines. Moreover, we clarify the connection between exercise, myokines and disease development, which may contribute to the understanding of a potential mechanism by which physical inactivity affects the process of metabolic diseases via myokines. Based on the current findings, myokines are important factors that mediate the effect of skeletal muscle on other organ functions and whole-body metabolism.
Collapse
Affiliation(s)
- Wentao Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| |
Collapse
|
19
|
Kim H, Cho SC, Jeong HJ, Lee HY, Jeong MH, Pyun JH, Ryu D, Kim M, Lee YS, Kim MS, Park SC, Lee YI, Kang JS. Indoprofen prevents muscle wasting in aged mice through activation of PDK1/AKT pathway. J Cachexia Sarcopenia Muscle 2020; 11:1070-1088. [PMID: 32096917 PMCID: PMC7432593 DOI: 10.1002/jcsm.12558] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/15/2020] [Accepted: 01/30/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Muscle wasting, resulting from aging or pathological conditions, leads to reduced quality of life, increased morbidity, and increased mortality. Much research effort has been focused on the development of exercise mimetics to prevent muscle atrophy and weakness. In this study, we identified indoprofen from a screen for peroxisome proliferator-activated receptor γ coactivator α (PGC-1α) inducers and report its potential as a drug for muscle wasting. METHODS The effects of indoprofen treatment on dexamethasone-induced atrophy in mice and in 3-phosphoinositide-dependent protein kinase-1 (PDK1)-deleted C2C12 myotubes were evaluated by immunoblotting to determine the expression levels of myosin heavy chain and anabolic-related and oxidative metabolism-related proteins. Young, old, and disuse-induced muscle atrophic mice were administered indoprofen (2 mg/kg body weight) by gavage. Body weight, muscle weight, grip strength, isometric force, and muscle histology were assessed. The expression levels of muscle mass-related and function-related proteins were analysed by immunoblotting or immunostaining. RESULTS In young (3-month-old) and aged (22-month-old) mice, indoprofen treatment activated oxidative metabolism-related enzymes and led to increased muscle mass. Mechanistic analysis using animal models and muscle cells revealed that indoprofen treatment induced the sequential activation of AKT/p70S6 kinase (S6K) and AMP-activated protein kinase (AMPK), which in turn can augment protein synthesis and PGC-1α induction, respectively. Structural prediction analysis identified PDK1 as a target of indoprofen and, indeed, short-term treatment with indoprofen activated the PDK1/AKT/S6K pathway in muscle cells. Consistent with this finding, PDK1 inhibition abrogated indoprofen-induced AKT/S6K activation and hypertrophic response. CONCLUSIONS Our findings demonstrate the effects of indoprofen in boosting skeletal muscle mass through the sequential activation of PDK1/AKT/S6K and AMPK/PGC-1α. Taken together, our results suggest that indoprofen represents a potential drug to prevent muscle wasting and weakness related to aging or muscle diseases.
Collapse
Affiliation(s)
- Hyebeen Kim
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, South Korea
| | - Sung Chun Cho
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Hyeon-Ju Jeong
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, South Korea
| | - Hye-Young Lee
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, South Korea
| | - Myong-Ho Jeong
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, South Korea
| | - Jung-Hoon Pyun
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, South Korea
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, South Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, South Korea
| | - MinSeok Kim
- School of Undergraduate Studies, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Young-Sam Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Minseok S Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Sang Chul Park
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Yun-Il Lee
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, South Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
20
|
Arneson-Wissink PC, Hogan KA, Ducharme AM, Samani A, Jatoi A, Doles JD. The wasting-associated metabolite succinate disrupts myogenesis and impairs skeletal muscle regeneration. JCSM RAPID COMMUNICATIONS 2020; 3:56-69. [PMID: 32905522 PMCID: PMC7470228 DOI: 10.1002/rco2.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND Muscle wasting is a debilitating co-morbidity affecting most advanced cancer patients. Alongside enhanced muscle catabolism, defects in muscle repair/regeneration contribute to cancer-associated wasting. Among the factors implicated in suppression of muscle regeneration are cytokines that interfere with myogenic signal transduction pathways. Less understood is how other cancer/wasting-associated cues, such as metabolites, contribute to muscle dysfunction. This study investigates how the metabolite succinate affects myogenesis and muscle regeneration. METHODS We leveraged an established ectopic metabolite treatment (cell permeable dimethyl-succinate) strategy to evaluate the ability of intracellular succinate elevation to 1) affect myoblast homeostasis (proliferation, apoptosis), 2) disrupt protein dynamics and induce wasting-associated atrophy, and 3) modulate in vitro myogenesis. In vivo succinate supplementation experiments (2% succinate, 1% sucrose vehicle) were used to corroborate and extend in vitro observations. Metabolic profiling and functional metabolic studies were then performed to investigate the impact of succinate elevation on mitochondria function. RESULTS We found that in vitro succinate supplementation elevated intracellular succinate about 2-fold, and did not have an impact on proliferation or apoptosis of C2C12 myoblasts. Elevated succinate had minor effects on protein homeostasis (~25% decrease in protein synthesis assessed by OPP staining), and no significant effect on myotube atrophy. Succinate elevation interfered with in vitro myoblast differentiation, characterized by significant decreases in late markers of myogenesis and fewer nuclei per myosin heavy chain positive structure (assessed by immunofluorescence staining). While mice orally administered succinate did not exhibit changes in overall body composition or whole muscle weights, these mice displayed smaller muscle myofiber diameters (~6% decrease in the mean of non-linear regression curves fit to the histograms of minimum feret diameter distribution), which was exacerbated when muscle regeneration was induced with barium chloride injury. Significant decreases in the mean of non-linear regression curves fit to the histograms of minimum feret diameter distributions were observed 7 days and 28 days post injury. Elevated numbers of myogenin positive cells (3-fold increase) supportive of the differentiation defects observed in vitro were observed 28 days post injury. Metabolic profiling and functional metabolic assessment of myoblasts revealed that succinate elevation caused both widespread metabolic changes and significantly lowered maximal cellular respiration (~35% decrease). CONCLUSIONS This study broadens the repertoire of wasting-associated factors that can directly modulate muscle progenitor cell function and strengthens the hypothesis that metabolic derangements are significant contributors to impaired muscle regeneration, an important aspect of cancer-associated muscle wasting.
Collapse
Affiliation(s)
- Paige C Arneson-Wissink
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Kelly A Hogan
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Alexandra M Ducharme
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Adrienne Samani
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Aminah Jatoi
- Department of Oncology, Mayo Clinic, Rochester,
Minnesota
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
- Corresponding Author: Jason D Doles, Department of
Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Guggenheim
16-11A1, Rochester, MN 55905, Tel: (507) 284-9372, Fax: (507) 284-3383,
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW This review summarizes the important role that metabolism plays in driving maturation of human pluripotent stem cell-derived cardiomyocytes. RECENT FINDINGS Human pluripotent stem cell-derived cardiomyocytes provide a model system for human cardiac biology. However, these models have been unable to fully recapitulate the maturity observed in the adult heart. By simulating the glucose to fatty acid transition observed in neonatal mammals, human pluripotent stem cell-derived cardiomyocytes undergo structural and functional maturation also accompanied by transcriptional changes and cell cycle arrest. The role of metabolism in energy production, signaling, and epigenetic modifications illustrates that metabolism and cellular phenotype are intimately linked. Further understanding of key metabolic factors driving cardiac maturation will facilitate the generation of more mature human pluripotent stem cell-derived cardiomyocyte models. This will increase our understanding of cardiac biology and potentially lead to novel therapeutics to enhance heart function.
Collapse
|
22
|
Knudsen JR, Li Z, Persson KW, Li J, Henriquez-Olguin C, Jensen TE. Contraction-regulated mTORC1 and protein synthesis: Influence of AMPK and glycogen. J Physiol 2020; 598:2637-2649. [PMID: 32372406 DOI: 10.1113/jp279780] [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: 03/08/2020] [Accepted: 04/14/2020] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS AMP-activated protein kinase (AMPK)-dependent Raptor Ser792 phosphorylation does not influence mechanistic target of rapamycin complex 1 (mTORC1)-S6K1 activation by intense muscle contraction. α2 -AMPK activity-deficient mice have lower contraction-stimulated protein synthesis. Increasing glycogen activates mTORC1-S6K1. Normalizing muscle glycogen content rescues reduced protein synthesis in AMPK-deficient mice. ABSTRACT The mechansitic target of rapamycin complex 1 (mTORC1)-S6K1 signalling pathway regulates muscle growth-related protein synthesis and is antagonized by AMP-activated protein kinase (AMPK) in multiple cell types. Resistance exercise stimulates skeletal muscle mTORC1-S6K1 and AMPK signalling and post-contraction protein synthesis. Glycogen inhibits AMPK and has been proposed as a pro-anabolic stimulus. The present study aimed to investigate how muscle mTORC1-S6K1 signalling and protein synthesis respond to resistance exercise-mimicking contraction in the absence of AMPK and with glycogen manipulation. Resistance exercise-mimicking unilateral in situ contraction of musculus quadriceps femoris in anaesthetized wild-type and dominant negative α2 AMPK kinase dead transgenic (KD-AMPK) mice, measuring muscle mTORC1 and AMPK signalling immediately (0 h) and 4 h post-contraction, and protein-synthesis at 4 h. Muscle glycogen manipulation by 5 day oral gavage of the glycogen phosphorylase inhibitor CP316819 and sucrose (80 g L-1 ) in the drinking water prior to in situ contraction. The mTORC1-S6K1 and AMPK signalling axes were coactivated immediately post-contraction, despite potent AMPK-dependent Ser792 phosphorylation on the mTORC1 subunit raptor. KD-AMPK muscles displayed normal mTORC1-S6K1 activation at 0 h and 4 h post-exercise, although there was impaired contraction-stimulated protein synthesis 4 h post-contraction. Pharmacological/dietary elevation of muscle glycogen content augmented contraction-stimulated mTORC1-S6K1-S6 signalling and rescued the reduced protein synthesis-response in KD-AMPK to wild-type levels. mTORC-S6K1 signalling is not influenced by α2 -AMPK during or after intense muscle contraction. Elevated glycogen augments mTORC1-S6K1 signalling. α2 -AMPK-deficient KD-AMPK mice display impaired contraction-induced muscle protein synthesis, which can be rescued by normalizing muscle glycogen content.
Collapse
Affiliation(s)
- Jonas R Knudsen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Zhencheng Li
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kaspar W Persson
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jingwen Li
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Carlos Henriquez-Olguin
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Mehra L, Bhattacharya A, Rawat H, Kumar A, Jaimini A, Mittal G. In-vitro and in-vivo functional observation studies to establish therapeutic potential of alpha-ketoglutarate against methotrexate induced liver injury. Biomed J 2020; 44:611-619. [PMID: 34736875 PMCID: PMC8640558 DOI: 10.1016/j.bj.2020.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/04/2020] [Accepted: 05/04/2020] [Indexed: 11/25/2022] Open
Abstract
Background Methotrexate (MTX) is widely used in chemotherapy but its associated hepatotoxicity is a major complication, limiting its use. This study evaluates possible therapeutic effect of oral alpha-ketoglutarate (AKG) supplementation against MTX-induced hepatotoxicity. Methods HepG2 cells were used to evaluate in-vitro cyto-protection conferred by AKG against MTX induced cytotoxicity. For in-vivo animal study, rats were divided into three groups. Group-I served as control. Group-II animals were administered single intraperitoneal injection of MTX (20 mg/kg/body weight), while Group-III received MTX as in group-II followed by oral AKG (2 gm/kg body weight) for 5 days. 99mTc-Mebrofenin hepatobiliary study was performed under a gamma camera to determine real time functional status of rats’ livers. Multiple parameters concerning hepatic mebrofenin uptake and excretion, including Tpeak and T1/2 peak in control and treated animals were determined. Biochemical analysis of the liver homogenate in terms of hepatic enzyme activities in serum, antioxidant status, tissue factor activity, tissue collagen content and histological analysis of the liver tissue were also done. Results AKG supplementation significantly reversed MTX induced derangement in activities of serum liver enzymes [ALT and ALP (p = 0.003); AST (p = 0.005)], antioxidant status [LPO and GSH (p = 0.005); CAT (p = 0.004)], tissue factor activity (p = 0.005) and tissue collagen content (p = 0.005). Functional imaging confirmed that hepatic retention and fractional biliary excretion were significantly abnormal in MTX treated group (Tpeak: 234 s ± 40 s; T1/2 peak: 846sec ± 32sec) as compared to AKG supplemented group (Tpeak: 144 s ± 35sec; T1/2peak: 468sec ± 27sec). Hepatic extraction fraction (HEF) was 92.2 ± 1.8%, 48.7 ± 2.6% and 69.8 ± 4.3% in control, MTX and AKG supplemented rats respectively. Conclusion 99mTc-mebrofenin imaging strongly suggests therapeutic action of AKG in protecting liver damage by MTX in rats. Functional imaging parameters correlated well with biochemical and histopathological findings.
Collapse
Affiliation(s)
- Lalita Mehra
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | | | - Harish Rawat
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | - Amit Kumar
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | - Abhinav Jaimini
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | - Gaurav Mittal
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Delhi, India.
| |
Collapse
|
24
|
Yuan Y, Xu P, Jiang Q, Cai X, Wang T, Peng W, Sun J, Zhu C, Zhang C, Yue D, He Z, Yang J, Zeng Y, Du M, Zhang F, Ibrahimi L, Schaul S, Jiang Y, Wang J, Sun J, Wang Q, Liu L, Wang S, Wang L, Zhu X, Gao P, Xi Q, Yin C, Li F, Xu G, Zhang Y, Shu G. Exercise-induced α-ketoglutaric acid stimulates muscle hypertrophy and fat loss through OXGR1-dependent adrenal activation. EMBO J 2020; 39:e103304. [PMID: 32104923 PMCID: PMC7110140 DOI: 10.15252/embj.2019103304] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/25/2020] [Accepted: 01/28/2020] [Indexed: 12/24/2022] Open
Abstract
Beneficial effects of resistance exercise on metabolic health and particularly muscle hypertrophy and fat loss are well established, but the underlying chemical and physiological mechanisms are not fully understood. Here, we identified a myometabolite‐mediated metabolic pathway that is essential for the beneficial metabolic effects of resistance exercise in mice. We showed that substantial accumulation of the tricarboxylic acid cycle intermediate α‐ketoglutaric acid (AKG) is a metabolic signature of resistance exercise performance. Interestingly, human plasma AKG level is also negatively correlated with BMI. Pharmacological elevation of circulating AKG induces muscle hypertrophy, brown adipose tissue (BAT) thermogenesis, and white adipose tissue (WAT) lipolysis in vivo. We further found that AKG stimulates the adrenal release of adrenaline through 2‐oxoglutarate receptor 1 (OXGR1) expressed in adrenal glands. Finally, by using both loss‐of‐function and gain‐of‐function mouse models, we showed that OXGR1 is essential for AKG‐mediated exercise‐induced beneficial metabolic effects. These findings reveal an unappreciated mechanism for the salutary effects of resistance exercise, using AKG as a systemically derived molecule for adrenal stimulation of muscle hypertrophy and fat loss.
Collapse
Affiliation(s)
- Yexian Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Qingyan Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xingcai Cai
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tao Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wentong Peng
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiajie Sun
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Canjun Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Cha Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Dong Yue
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhihui He
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jinping Yang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yuxian Zeng
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Man Du
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fenglin Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lucas Ibrahimi
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Sarah Schaul
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, USA
| | - Jiqiu Wang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Sun
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaoping Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University Guangzhou, Guangzhou, China
| | - Liming Liu
- State Key Laboratory of Food Science and Technology and Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi, China
| | - Songbo Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lina Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaotong Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ping Gao
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianyun Xi
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Cong Yin
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fan Li
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Guli Xu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yongliang Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
25
|
Kirchberg FF, Hellmuth C, Totzauer M, Uhl O, Closa-Monasterolo R, Escribano J, Gruszfeld D, Gradowska K, Verduci E, Mariani B, Moretti M, Rousseaux D, Koletzko B. Impact of infant protein supply and other early life factors on plasma metabolome at 5.5 and 8 years of age: a randomized trial. Int J Obes (Lond) 2020; 44:69-81. [PMID: 31300705 PMCID: PMC7617055 DOI: 10.1038/s41366-019-0398-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 01/06/2019] [Accepted: 04/19/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVES A high dairy protein intake in infancy, maternal pre-pregnancy BMI, and delivery mode are documented early programming factors that modulate the later risk of obesity and other health outcomes, but the mechanisms of action are not understood. METHODS The Childhood Obesity Project is a European multicenter, double-blind, randomized clinical trial that enrolled healthy infants. Participating infants were either breastfed (BF) or randomized to receive higher (HP) or lower protein (LP) content formula in the first year of life. At the ages 5.5 years (n = 276) and 8 years (n = 232), we determined plasma metabolites by liquid chromatography tandem-mass-spectrometry of which 226 and 185 passed quality control at 5.5 years and 8 years, respectively. We assessed the effects of infant feeding, maternal pre-pregnancy BMI, smoking in pregnancy, delivery mode, parity, birth weight and length, and weight gain (0-24 months) on the metabolome at 5.5 and 8 years. RESULTS At 5.5 years, plasma alpha-ketoglutarate and the acylcarnitine/BCAA ratios tended to be higher in the HP than in the LP group, but no metabolite reached statistical significance (Pbonferroni>0.09). There were no group differences at 8 years. Quantification of the impact of early programming factors revealed that the intervention group explained 0.6% of metabolome variance at both time points. Except for country of residence that explained 16% and 12% at 5.5 years and 8 years, respectively, none of the other factors explained considerably more variance than expected by chance. CONCLUSIONS Plasma metabolome was largely unaffected by feeding choice and other early programming factors and we could not prove the existence of a long term programming effect of the plasma metabolome.
Collapse
Affiliation(s)
- Franca Fabiana Kirchberg
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, 80337, Munich, Germany
| | - Christian Hellmuth
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, 80337, Munich, Germany
| | - Martina Totzauer
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, 80337, Munich, Germany
| | - Olaf Uhl
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, 80337, Munich, Germany
| | - Ricardo Closa-Monasterolo
- Pediatric, Nutrition and Human Development Research Unit, Universitat Rovira i Virgili, IISPV; C/ Dr. Mallafrè Guasch, 4, 43005, Tarragona Reus, Spain
| | - Joaquin Escribano
- Pediatric, Nutrition and Human Development Research Unit, Universitat Rovira i Virgili, IISPV; C/ Dr. Mallafrè Guasch, 4, 43005, Tarragona Reus, Spain
| | - Dariusz Gruszfeld
- Neonatal Department, Children's Memorial Health Institute, aleja Dzieci Polskich 20; 04-730 Warszawa, Polen, Warszawa, Poland
| | - Kinga Gradowska
- Neonatal Department, Children's Memorial Health Institute, aleja Dzieci Polskich 20; 04-730 Warszawa, Polen, Warszawa, Poland
| | - Elvira Verduci
- Department of Paediatrics, San Paolo Hospital, University of Milan, Via Antonio di Rudinì, 8, 20142, Milano, MI, Italy
| | - Benedetta Mariani
- Department of Paediatrics, San Paolo Hospital, University of Milan, Via Antonio di Rudinì, 8, 20142, Milano, MI, Italy
| | - Melissa Moretti
- CHC Sant Vincent, Rue François-Lefèbvre 207, 4000, Liège, Belgium
| | | | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, 80337, Munich, Germany.
| |
Collapse
|
26
|
FOXP1 inhibits high glucose-induced ECM accumulation and oxidative stress in mesangial cells. Chem Biol Interact 2019; 313:108818. [DOI: 10.1016/j.cbi.2019.108818] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/12/2019] [Accepted: 09/05/2019] [Indexed: 01/09/2023]
|
27
|
Zhao L, Guo H, Sun H. Effects of low-protein diet supplementation with alpha-ketoglutarate on growth performance, nitrogen metabolism and mTOR signalling pathway of skeletal muscle in piglets. J Anim Physiol Anim Nutr (Berl) 2019; 104:300-309. [PMID: 31674084 DOI: 10.1111/jpn.13230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/21/2019] [Accepted: 09/16/2019] [Indexed: 11/30/2022]
Abstract
Excessive protein levels in diets result in incomplete digestion of nitrogenous nutrients that are excreted from the body, causing environment pollution. Alpha-ketoglutarate (AKG) has been reported to decrease dietary protein levels, promote intestinal health in piglets and reduce environmental pollution. However, the underlying mechanisms of AKG are largely unknown. The objective of this study was to determine the effects of low-protein diet supplementation of AKG on the growth performance, nitrogen metabolism, relative expression of amino acid transporter genes and mTOR signalling pathway of skeletal muscle in piglets. Forty-eight piglets with an initial weight of 11.53 ± 0.04 kg were randomly divided into four groups. Each group had four replicates, and each replicate had three pigs. A low-protein (LP) diet (crude protein was 14.96%) served as the control without AKG, while 0.5%, 1.0% and 1.5% AKG were added to the LP diet for the other experimental groups. The trial period lasted for 28 days. Compared with the LP group, the LP + 1.0%A and LP + 1.5%A groups increased the growth performance (p < .05);increased the mRNA levels of amino acid transporters in the duodenum, anterior jejunum and posterior jejunum (p < .05); and reduced faecal nitrogen and urine nitrogen emissions (p < .05). They also showed greater mRNA levels and phosphorylated protein levels for S6 kinase beta (S6K) (p < .05), mammalian target of rapamycin (mTOR) (p < .05) and 4E-binding protein 1 (4EBP1) (p < .05) in skeletal muscle. An LP diet supplemented with AKG activated the mTOR signalling and promoted the ability of the small intestine to absorb protein, thereby increasing protein deposition. Taken together, an LP diet supplemented with AKG provides a theoretical basis for the promotion and application of AKG in piglet production.
Collapse
Affiliation(s)
- Lei Zhao
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, China.,Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China.,Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Haolu Guo
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, China.,Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China.,Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Hui Sun
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, China.,Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China.,Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| |
Collapse
|
28
|
Ozaki M. Cellular and molecular mechanisms of liver regeneration: Proliferation, growth, death and protection of hepatocytes. Semin Cell Dev Biol 2019; 100:62-73. [PMID: 31669133 DOI: 10.1016/j.semcdb.2019.10.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 01/08/2023]
Abstract
Liver regeneration is an important and necessary process that the liver depends on for recovery from injury. The regeneration process consists of a complex network of cells and organs, including liver cells (parenchymal and non-parenchymal cells) and extrahepatic organs (thyroid, adrenal glands, pancreas, duodenum, spleen, and autonomic nervous system). The regeneration process of a normal, healthy liver depends mainly on hepatocyte proliferation, growth, and programmed cell death. Cell proliferation and growth are regulated in a cooperative manner by interleukin (IL)-6/janus kinase (Jak)/signal transducers and activators of transcription-3 (STAT3), and phosphoinositide 3-kinase (PI3-K)/phosphoinositide-dependent protein kinase 1 (PDK1)/Akt pathways. The IL-6/Jak/STAT3 pathway regulates hepatocyte proliferation and protects against cell death and oxidative stress. The PI3-K/PDK1/Akt pathway is primarily responsible for the regulation of cell size, sending mitotic signals in addition to pro-survival, antiapoptotic and antioxidative signals. Though programmed cell death may interfere with liver regeneration in a pathological situation, it seems to play an important role during the termination phase, even in a normal, healthy liver regeneration. However, further study is needed to fully elucidate the mechanisms regulating the processes of liver regeneration with regard to cell-to-cell and organ-to-organ networks at the molecular and cellular levels.
Collapse
Affiliation(s)
- Michitaka Ozaki
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido University, N12, W5, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan.
| |
Collapse
|
29
|
Wang J, Li Y, Luo P, Chen Y, Xi Q, Wu H, Zhao W, Shu G, Wang S, Gao P, Zhu X, Zhang Y, Jiang Q, Wang L. Oral supplementation with ginseng polysaccharide promotes food intake in mice. Brain Behav 2019; 9:e01340. [PMID: 31392839 PMCID: PMC6749478 DOI: 10.1002/brb3.1340] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Ginseng polysaccharide (GPS, same as Panax polysaccharide) is a kind of polysaccharide extracted from ginseng. It has been reported that GPS has the ability to activate innate immunity, regulates blood sugar balance, and improves antioxidant capacity, but the effect on feeding behavior and its mechanism remains unclear. METHOD To investigate the possible effect of GPS on feeding behavior of animals, mice were supplied with GPS in water, and food intake, hedonic feeding behavior, anxiety-like behavior, expression of appetite-regulation peptides in the central nervous system and glucose-related hormone levels in the serum of mice were measured. RESULTS Ginseng polysaccharide significantly increased the average daily food intake in mice and promoted hedonic eating behavior. Meanwhile, the levels of serum glucose and glucagon were significantly reduced by GPS, and GPS promoted hypothalamic neuropeptide Y expression, inhibited proopiomelanocortin (POMC) expression, and reduced dopamine D1 receptor (DRD1) levels in the midbrain. We also found that the anxiety level of mice was significantly lower after GPS intake. In conclusion, oral supplementation with GPS promoted food intake in mice, most likely through the regulation of circulating glucose levels.
Collapse
Affiliation(s)
- Jiawen Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Yongxiang Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Pei Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Yuhuang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Hanyu Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Weijie Zhao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.,National Engineering Research Center for the Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
30
|
Wang T, Xu Y, Yuan Y, Xu P, Zhang C, Li F, Wang L, Yin C, Zhang L, Cai X, Zhu C, Xu J, Liang B, Schaul S, Xie P, Yue D, Liao Z, Yu L, Luo L, Zhou G, Yang J, He Z, Du M, Zhou Y, Deng B, Wang S, Gao P, Zhu X, Xi Q, Zhang Y, Shu G, Jiang Q. Succinate induces skeletal muscle fiber remodeling via SUNCR1 signaling. EMBO Rep 2019; 20:e47892. [PMID: 31318145 PMCID: PMC6727026 DOI: 10.15252/embr.201947892] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/13/2019] [Accepted: 06/26/2019] [Indexed: 01/08/2023] Open
Abstract
The conversion of skeletal muscle fiber from fast twitch to slow-twitch is important for sustained and tonic contractile events, maintenance of energy homeostasis, and the alleviation of fatigue. Skeletal muscle remodeling is effectively induced by endurance or aerobic exercise, which also generates several tricarboxylic acid (TCA) cycle intermediates, including succinate. However, whether succinate regulates muscle fiber-type transitions remains unclear. Here, we found that dietary succinate supplementation increased endurance exercise ability, myosin heavy chain I expression, aerobic enzyme activity, oxygen consumption, and mitochondrial biogenesis in mouse skeletal muscle. By contrast, succinate decreased lactate dehydrogenase activity, lactate production, and myosin heavy chain IIb expression. Further, by using pharmacological or genetic loss-of-function models generated by phospholipase Cβ antagonists, SUNCR1 global knockout, or SUNCR1 gastrocnemius-specific knockdown, we found that the effects of succinate on skeletal muscle fiber-type remodeling are mediated by SUNCR1 and its downstream calcium/NFAT signaling pathway. In summary, our results demonstrate succinate induces transition of skeletal muscle fiber via SUNCR1 signaling pathway. These findings suggest the potential beneficial use of succinate-based compounds in both athletic and sedentary populations.
Collapse
Affiliation(s)
- Tao Wang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Ya‐Qiong Xu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Ye‐Xian Yuan
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Ping‐Wen Xu
- Division of EndocrinologyDepartment of MedicineThe University of Illinois at ChicagoChicagoILUSA
| | - Cha Zhang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Fan Li
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Li‐Na Wang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Cong Yin
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Lin Zhang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Xing‐Cai Cai
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Can‐Jun Zhu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Jing‐Ren Xu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Bing‐Qing Liang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Sarah Schaul
- Division of EndocrinologyDepartment of MedicineThe University of Illinois at ChicagoChicagoILUSA
| | - Pei‐Pei Xie
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Dong Yue
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Zheng‐Rui Liao
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Lu‐Lu Yu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Lv Luo
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Gan Zhou
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Jin‐Ping Yang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Zhi‐Hui He
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Man Du
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Yu‐Ping Zhou
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Bai‐Chuan Deng
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Song‐Bo Wang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Ping Gao
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Xiao‐Tong Zhu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Qian‐Yun Xi
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Yong‐Liang Zhang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Gang Shu
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
- National Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| | - Qing‐Yan Jiang
- Guangdong Province Key Laboratory of Animal Nutritional RegulationCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
- National Engineering Research Center for Breeding Swine IndustryCollege of Animal ScienceSouth China Agricultural UniversityGuangzhouGuangdongChina
| |
Collapse
|
31
|
The multifaceted contribution of α-ketoglutarate to tumor progression: An opportunity to exploit? Semin Cell Dev Biol 2019; 98:26-33. [PMID: 31175937 DOI: 10.1016/j.semcdb.2019.05.031] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 01/25/2023]
Abstract
The thriving field that constitutes cancer metabolism has unveiled some groundbreaking facts over the past two decades, at the heart of which is the TCA cycle and its intermediates. As such and besides its metabolic role, α-ketoglutarate was shown to withstand a wide range of physiological reactions from protection against oxidative stress, collagen and bone maintenance to development and immunity. Most importantly, it constitutes the rate-limiting substrate of 2-oxoglutarate-dependent dioxygenases family enzymes, which are involved in hypoxia sensing and in the shaping of cellular epigenetic landscape, two major drivers of oncogenic transformation. Based on literature reports, we hereby review the benefits of this metabolite as a possible novel adjuvant therapeutic opportunity to target tumor progression. This article is part of the special issue "Mitochondrial metabolic alterations in cancer cells and related therapeutic targets".
Collapse
|
32
|
Baracco EE, Castoldi F, Durand S, Enot DP, Tadic J, Kainz K, Madeo F, Chery A, Izzo V, Maiuri MC, Pietrocola F, Kroemer G. α-Ketoglutarate inhibits autophagy. Aging (Albany NY) 2019; 11:3418-3431. [PMID: 31173576 PMCID: PMC6594794 DOI: 10.18632/aging.102001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/24/2019] [Indexed: 12/20/2022]
Abstract
The metabolite α-ketoglutarate is membrane-impermeable, meaning that it is usually added to cells in the form of esters such as dimethyl -ketoglutarate (DMKG), trifluoromethylbenzyl α-ketoglutarate (TFMKG) and octyl α-ketoglutarate (O-KG). Once these compounds cross the plasma membrane, they are hydrolyzed by esterases to generate α-ketoglutarate, which remains trapped within cells. Here, we systematically compared DMKG, TFMKG and O-KG for their metabolic and functional effects. All three compounds similarly increased the intracellular levels of α-ketoglutarate, yet each of them had multiple effects on other metabolites that were not shared among the three agents, as determined by mass spectrometric metabolomics. While all three compounds reduced autophagy induced by culture in nutrient-free conditions, TFMKG and O-KG (but not DMKG) caused an increase in baseline autophagy in cells cultured in complete medium. O-KG (but neither DMKG nor TFMK) inhibited oxidative phosphorylation and exhibited cellular toxicity. Altogether, these results support the idea that intracellular α-ketoglutarate inhibits starvation-induced autophagy and that it has no direct respiration-inhibitory effect.
Collapse
Affiliation(s)
- Elisa Elena Baracco
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Francesca Castoldi
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sylvère Durand
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - David P. Enot
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jelena Tadic
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, Austria
| | - Katharina Kainz
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, Austria
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Alexis Chery
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Valentina Izzo
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Federico Pietrocola
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Institute for Research in Biomedicine, Barcelona, Spain
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Descartes, Université Paris Diderot, "Metabolism, Cancer and Immunity", Paris 75006, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
33
|
Valenzuela PL, Morales JS, Emanuele E, Pareja-Galeano H, Lucia A. Supplements with purported effects on muscle mass and strength. Eur J Nutr 2019; 58:2983-3008. [PMID: 30604177 DOI: 10.1007/s00394-018-1882-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Several supplements are purported to promote muscle hypertrophy and strength gains in healthy subjects, or to prevent muscle wasting in atrophying situations (e.g., ageing or disuse periods). However, their effectiveness remains unclear. METHODS This review summarizes the available evidence on the beneficial impacts of several popular supplements on muscle mass or strength. RESULTS Among the supplements tested, nitrate and caffeine returned sufficient evidence supporting their acute beneficial effects on muscle strength, whereas the long-term consumption of creatine, protein and polyunsaturated fatty acids seems to consistently increase or preserve muscle mass and strength (evidence level A). On the other hand, mixed or unclear evidence was found for several popular supplements including branched-chain amino acids, adenosine triphosphate, citrulline, β-Hydroxy-β-methylbutyrate, minerals, most vitamins, phosphatidic acid or arginine (evidence level B), weak or scarce evidence was found for conjugated linoleic acid, glutamine, resveratrol, tribulus terrestris or ursolic acid (evidence level C), and no evidence was found for other supplements such as ornithine or α-ketoglutarate (evidence D). Of note, although most supplements appear to be safe when consumed at typical doses, some adverse events have been reported for some of them (e.g., caffeine, vitamins, α-ketoglutarate, tribulus terrestris, arginine) after large intakes, and there is insufficient evidence to determine the safety of many frequently used supplements (e.g., ornithine, conjugated linoleic acid, ursolic acid). CONCLUSION In summary, despite their popularity, there is little evidence supporting the use of most supplements, and some of them have been even proven ineffective or potentially associated with adverse effects.
Collapse
Affiliation(s)
- Pedro L Valenzuela
- Department of Sport and Health, Spanish Agency for Health Protection in Sport (AEPSAD), Madrid, Spain.,Physiology Unit. Systems Biology Department, University of Alcalá, Madrid, Spain
| | - Javier S Morales
- Faculty of Sport Sciences, Universidad Europea De Madrid, Villaviciosa De Odón, 28670, Madrid, Spain
| | | | - Helios Pareja-Galeano
- Faculty of Sport Sciences, Universidad Europea De Madrid, Villaviciosa De Odón, 28670, Madrid, Spain. .,Research Institute of the Hospital 12 De Octubre (i+12), Madrid, Spain.
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea De Madrid, Villaviciosa De Odón, 28670, Madrid, Spain.,Research Institute of the Hospital 12 De Octubre (i+12), Madrid, Spain
| |
Collapse
|
34
|
Wackerhage H, Schoenfeld BJ, Hamilton DL, Lehti M, Hulmi JJ. Stimuli and sensors that initiate skeletal muscle hypertrophy following resistance exercise. J Appl Physiol (1985) 2018; 126:30-43. [PMID: 30335577 DOI: 10.1152/japplphysiol.00685.2018] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
One of the most striking adaptations to exercise is the skeletal muscle hypertrophy that occurs in response to resistance exercise. A large body of work shows that a mammalian target of rapamycin complex 1 (mTORC1)-mediated increase of muscle protein synthesis is the key, but not sole, mechanism by which resistance exercise causes muscle hypertrophy. While much of the hypertrophy signaling cascade has been identified, the initiating, resistance exercise-induced and hypertrophy-stimulating stimuli have remained elusive. For the purpose of this review, we define an initiating, resistance exercise-induced and hypertrophy-stimulating signal as "hypertrophy stimulus," and the sensor of such a signal as "hypertrophy sensor." In this review we discuss our current knowledge of specific mechanical stimuli, damage/injury-associated and metabolic stress-associated triggers, as potential hypertrophy stimuli. Mechanical signals are the prime hypertrophy stimuli candidates, and a filamin-C-BAG3-dependent regulation of mTORC1, Hippo, and autophagy signaling is a plausible albeit still incompletely characterized hypertrophy sensor. Other candidate mechanosensing mechanisms are nuclear deformation-initiated signaling or several mechanisms related to costameres, which are the functional equivalents of focal adhesions in other cells. While exercise-induced muscle damage is probably not essential for hypertrophy, it is still unclear whether and how such muscle damage could augment a hypertrophic response. Interventions that combine blood flow restriction and especially low load resistance exercise suggest that resistance exercise-regulated metabolites could be hypertrophy stimuli, but this is based on indirect evidence and metabolite candidates are poorly characterized.
Collapse
Affiliation(s)
- Henning Wackerhage
- Department of Sport and Exercise Sciences, Technical University of Munich , Munich , Germany
| | | | - D Lee Hamilton
- Faculty of Health, School of Exercise and Nutrition Sciences, Deakin University , Victoria , Australia
| | - Maarit Lehti
- LIKES Research Centre for Physical Activity and Health , Jyväskylä , Finland
| | - Juha J Hulmi
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä , Jyväskylä , Finland
| |
Collapse
|
35
|
Wang Y, Ma J, Qiu W, Zhang J, Feng S, Zhou X, Wang X, Jin L, Long K, Liu L, Xiao W, Tang Q, Zhu L, Jiang Y, Li X, Li M. Guanidinoacetic Acid Regulates Myogenic Differentiation and Muscle Growth Through miR-133a-3p and miR-1a-3p Co-mediated Akt/mTOR/S6K Signaling Pathway. Int J Mol Sci 2018; 19:ijms19092837. [PMID: 30235878 PMCID: PMC6163908 DOI: 10.3390/ijms19092837] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 12/12/2022] Open
Abstract
Guanidinoacetic acid (GAA), an amino acid derivative that is endogenous to animal tissues including muscle and nerve, has been reported to enhance muscular performance. MicroRNA (miRNA) is a post-transcriptional regulator that plays a key role in nutrient-mediated myogenesis. However, the effects of GAA on myogenic differentiation and skeletal muscle growth, and the potential regulatory mechanisms of miRNA in these processes have not been elucidated. In this study, we investigated the effects of GAA on proliferation, differentiation, and growth in C2C12 cells and mice. The results showed that GAA markedly inhibited the proliferation of myoblasts, along with the down-regulation of cyclin D1 (CCND1) and cyclin dependent kinase 4 (CDK4) mRNA expression, and the upregulation of cyclin dependent kinase inhibitor 1A (P21) mRNA expression. We also demonstrated that GAA treatment stimulated myogenic differentiation 1 (MyoD) and myogenin (MyoG) mRNA expression, resulting in an increase in the myotube fusion rate. Meanwhile, GAA supplementation promoted myotube growth through increase in total myosin heavy chain (MyHC) protein level, myotubes thickness and gastrocnemius muscle cross-sectional area. Furthermore, small RNA sequencing revealed that a total of eight miRNAs, including miR-133a-3p and miR-1a-3p cluster, showed differential expression after GAA supplementation. To further study the function of miR-133a-3p and miR-1a-3p in GAA-induced skeletal muscle growth, we transfected miR-133a-3p and miR-1a-3p mimics into myotube, which also induced muscle growth. Through bioinformatics and a dual-luciferase reporter system, the target genes of miR-133a-3p and miR-1a-3p were determined. These two miRNAs were shown to modulate the Akt/mTOR/S6K signaling pathway by restraining target gene expression. Taken together, these findings suggest that GAA supplementation can promote myoblast differentiation and skeletal muscle growth through miR-133a-3p- and miR-1a-3p-induced activation of the AKT/mTOR/S6K signaling pathway.
Collapse
Affiliation(s)
- Yujie Wang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Jideng Ma
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Wanling Qiu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Jinwei Zhang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Siyuan Feng
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Xiankun Zhou
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Xun Wang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Long Jin
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Keren Long
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Lingyan Liu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Weihang Xiao
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Qianzi Tang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Li Zhu
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Yanzhi Jiang
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Xuewei Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| | - Mingzhou Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu 611130, China.
| |
Collapse
|
36
|
Chen J, Su W, Kang B, Jiang Q, Zhao Y, Fu C, Yao K. Supplementation with α-ketoglutarate to a low-protein diet enhances amino acid synthesis in tissues and improves protein metabolism in the skeletal muscle of growing pigs. Amino Acids 2018; 50:1525-1537. [PMID: 30167964 DOI: 10.1007/s00726-018-2618-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022]
Abstract
α-Ketoglutarate (AKG) is a crucial intermediate in the tricarboxylic acid (TCA) cycle and can be used for the production of ATP and amino acids in animal tissues. However, the effect of AKG on the expression patterns of genes involved in muscle protein metabolism is largely unknown, and the underlying mechanism remains to be elucidated. Therefore, we used young pigs to investigate the effects of a low crude protein (CP) diet and a low CP diet supplemented with AKG on protein accretion in their skeletal muscle. A total of 27 growing pigs with an initial body weight of 11.96 ± 0.18 kg were assigned randomly to one of the three diets: control (normal recommended 20% CP, NP), low CP (17% CP, LP), or low CP supplemented with 1% AKG (ALP). The pigs were fed their respective diets for 35 days. Free amino acid (AA) profile and hormone levels in the serum, and the expression of genes implicated in protein metabolism in skeletal muscle were examined. Results showed that compared with the control group or LP group, low-protein diets supplemented with AKG enhanced serum and intramuscular free AA concentrations, the mRNA abundances of AA transporters, and serum concentrations of insulin-like growth factor-1 (IGF-1), activated the mammalian target of rapamycin (mTOR) pathway, and decreased serum urea concentration and the mRNA levels for genes related to muscle protein degradation (P < 0.05). In conclusion, these results indicated that addition of AKG to a low-protein diet promotes amino acid synthesis in tissues and improves protein metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Jiashun Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China.,Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Wenxuan Su
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Baoju Kang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China
| | - Qian Jiang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Yurong Zhao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China
| | - Chenxing Fu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China. .,Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients and Hunan Collaborative Innovation Center of Animal Production Safety, Changsha, 410128, Hunan, China.
| | - Kang Yao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, China. .,Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center of Healthy Livestock, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China.
| |
Collapse
|
37
|
Cai X, Yuan Y, Liao Z, Xing K, Zhu C, Xu Y, Yu L, Wang L, Wang S, Zhu X, Gao P, Zhang Y, Jiang Q, Xu P, Shu G. α-Ketoglutarate prevents skeletal muscle protein degradation and muscle atrophy through PHD3/ADRB2 pathway. FASEB J 2018; 32:488-499. [PMID: 28939592 PMCID: PMC6266637 DOI: 10.1096/fj.201700670r] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022]
Abstract
Skeletal muscle atrophy due to excessive protein degradation is the main cause for muscle dysfunction, fatigue, and weakening of athletic ability. Endurance exercise is effective to attenuate muscle atrophy, but the underlying mechanism has not been fully investigated. α-Ketoglutarate (AKG) is a key intermediate of tricarboxylic acid cycle, which is generated during endurance exercise. Here, we demonstrated that AKG effectively attenuated corticosterone-induced protein degradation and rescued the muscle atrophy and dysfunction in a Duchenne muscular dystrophy mouse model. Interestingly, AKG also inhibited the expression of proline hydroxylase 3 (PHD3), one of the important oxidoreductases expressed under hypoxic conditions. Subsequently, we identified the β2 adrenergic receptor (ADRB2) as a downstream target for PHD3. We found AKG inhibited PHD3/ADRB2 interaction and therefore increased the stability of ADRB2. In addition, combining pharmacologic and genetic approaches, we showed that AKG rescues skeletal muscle atrophy and protein degradation through a PHD3/ADRB2 mediated mechanism. Taken together, these data reveal a mechanism for inhibitory effects of AKG on muscle atrophy and protein degradation. These findings not only provide a molecular basis for the potential use of exercise-generated metabolite AKG in muscle atrophy treatment, but also identify PHD3 as a potential target for the development of therapies for muscle wasting.-Cai, X., Yuan, Y., Liao, Z., Xing, K., Zhu, C., Xu, Y., Yu, L., Wang, L., Wang, S., Zhu, X., Gao, P., Zhang, Y., Jiang, Q., Xu, P., Shu, G. α-Ketoglutarate prevents skeletal muscle protein degradation and muscle atrophy through PHD3/ADRB2 pathway.
Collapse
MESH Headings
- Animals
- Corticosterone/pharmacology
- Disease Models, Animal
- Ketoglutaric Acids/therapeutic use
- Male
- Metabolic Networks and Pathways/drug effects
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Muscular Atrophy/prevention & control
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Procollagen-Proline Dioxygenase/metabolism
- Protein Stability/drug effects
- Proteolysis/drug effects
- Receptors, Adrenergic, beta-2/metabolism
Collapse
Affiliation(s)
- Xingcai Cai
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Yexian Yuan
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Zhengrui Liao
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Kongping Xing
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Yaqiong Xu
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Lulu Yu
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Xiaotong Zhu
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Pingwen Xu
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou, China;
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
38
|
Valdivieso P, Vaughan D, Laczko E, Brogioli M, Waldron S, Rittweger J, Flück M. The Metabolic Response of Skeletal Muscle to Endurance Exercise Is Modified by the ACE-I/D Gene Polymorphism and Training State. Front Physiol 2017; 8:993. [PMID: 29311951 PMCID: PMC5735290 DOI: 10.3389/fphys.2017.00993] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 11/20/2017] [Indexed: 01/13/2023] Open
Abstract
The insertion/deletion polymorphism in the gene for the regulator of vascular tone, angiotensin-converting enzyme (ACE), is the prototype of a genetic influence on physical fitness and this involves an influence on capillary supply lines and dependent aerobic metabolism in skeletal muscle. The respective interaction of ACE-I/D genotype and training status on local metabolic and angiogenic reactions in exercised muscle is not known. Toward this end we characterized the metabolomic and angiogenic response in knee extensor muscle, m. vastus lateralis, in 18 untrained and 34 endurance-trained (physically active, [Formula: see text]O2max > 50 mL min-1 kg-1) white British men to an exhaustive bout of one-legged cycling exercise. We hypothesized that training status and ACE-I/D genotype affect supply-related muscle characteristics of exercise performance in correspondence to ACE expression and angiotensin 2 levels. ACE-I/D genotype and training status developed an interaction effect on the cross-sectional area (CSA) of m. vastus lateralis and mean CSA of slow type fibers, which correlated with peak power output (r ≥ 0.44). Genotype × training interactions in muscle also resolved for exercise-induced alterations of 22 metabolites, 8 lipids, glycogen concentration (p = 0.016), ACE transcript levels (p = 0.037), and by trend for the pro-angiogenic factor tenascin-C post exercise (p = 0.064). Capillary density (p = 0.001), capillary-to-fiber ratio (p = 0.010), systolic blood pressure (p = 0.014), and exercise-induced alterations in the pro-angiogenic protein VEGF (p = 0.043) depended on the ACE-I/D genotype alone. Our observations indicate that variability in aerobic performance in the studied subjects was in part reflected by an ACE-I/D-genotype-modulated metabolic phenotype of a major locomotor muscle. Repeated endurance exercise appeared to override this genetic influence in skeletal muscle by altering the ACE-related metabolic response and molecular aspects of the angiogenic response to endurance exercise.
Collapse
Affiliation(s)
- Paola Valdivieso
- Laboratory for Muscle Plasticity, Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - David Vaughan
- The Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, United Kingdom
| | - Endre Laczko
- Functional Genomics Center Zurich, ETH, University of Zurich, Zurich, Switzerland
| | - Michael Brogioli
- The Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, United Kingdom
| | - Sarah Waldron
- The Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, United Kingdom
| | - Jörn Rittweger
- The Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, United Kingdom.,Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Martin Flück
- Laboratory for Muscle Plasticity, Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland.,The Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
39
|
Yuan Y, Xu Y, Xu J, Liang B, Cai X, Zhu C, Wang L, Wang S, Zhu X, Gao P, Wang X, Zhang Y, Jiang Q, Shu G. Succinate promotes skeletal muscle protein synthesis via Erk1/2 signaling pathway. Mol Med Rep 2017; 16:7361-7366. [PMID: 28944867 PMCID: PMC5865866 DOI: 10.3892/mmr.2017.7554] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/07/2017] [Indexed: 12/25/2022] Open
Abstract
It is well known that endurance training is effective to attenuate skeletal muscle atrophy. Succinate is a typical TCA metabolite, of which exercise could dramatically increase the content. The present study aimed to investigate the effect of succinate on protein synthesis in skeletal muscle, and try to delineate the underlying mechanism. The in vitro study revealed that succinate dose‑dependently increased protein synthesis in C2C12 myotube along with the enhancement of phosphorylation levels of AKT Serine/Threonine Kinase 1(Akt), mammalian target of rapamycin, S6, eukaryotic translation initiation factor 4E, 4E binding protein 1 and forkhead box O (FoxO) 3a. Furthermore, it was demonstrated that 20 mM succinate markedly increased [Ca2+]i. Then, the phospho‑extracellular regulated kinase (Erk), ‑Akt level and the crosstalk between Erk and Akt were elevated in response to succinate. Notably, the Erk antagonist (U0126) or mTOR inhibitor (rapamycin) abolished the effect of succinate on protein synthesis. The in vivo study verified that succinate dose‑dependently increased the protein synthesis, in addition to phosphorylation levels of Erk, Akt and FoxO3a in gastrocnemius muscle. In summary, these findings demonstrated that succinate promoted skeletal muscle protein deposition via Erk/Akt signaling pathway.
Collapse
Affiliation(s)
- Yexian Yuan
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Yaqiong Xu
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Jingren Xu
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Bingqing Liang
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Xingcai Cai
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Canjun Zhu
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Lina Wang
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Songbo Wang
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Xiaotong Zhu
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Ping Gao
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Xiuqi Wang
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Yongliang Zhang
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Qingyan Jiang
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| | - Gang Shu
- Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
- Animal Nutrition Control Research Alliance, South China Agricultural University, Guangzhou, Guangdong 510642, P.R. China
| |
Collapse
|
40
|
Ritterhoff J, Tian R. Metabolism in cardiomyopathy: every substrate matters. Cardiovasc Res 2017; 113:411-421. [PMID: 28395011 DOI: 10.1093/cvr/cvx017] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/01/2017] [Indexed: 12/12/2022] Open
Abstract
Cardiac metabolism is highly adaptive to changes in fuel availability and the energy demand of the heart. This metabolic flexibility is key for the heart to maintain its output during the development and in response to stress. Alterations in substrate preference have been observed in multiple disease states; a clear understanding of their impact on cardiac function in the long term is critical for the development of metabolic therapies. In addition, the contribution of cellular metabolism to growth, survival, and other signalling pathways through the generation of metabolic intermediates has been increasingly noted, adding another layer of complexity to the impact of metabolism on cardiac function. In a quest to understand the complexity of the cardiac metabolic network, genetic tools have been engaged to manipulate cardiac metabolism in a variety of mouse models. The ability to engineer cardiac metabolism in vivo has provided tremendous insights and brought about conceptual innovations. In this review, we will provide an overview of the cardiac metabolic network and highlight alterations observed during cardiac development and pathological hypertrophy. We will focus on consequences of altered substrate preference on cardiac response to chronic stresses through energy providing and non-energy providing pathways.
Collapse
|
41
|
Tang L, Zhang J, Zhao X, Li N, Jian W, Sun S, Guo J, Sun L, Ta D. Low-Intensity Pulsed Ultrasound Promotes Exercise-Induced Muscle Hypertrophy. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:1411-1420. [PMID: 28461063 DOI: 10.1016/j.ultrasmedbio.2017.02.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/08/2017] [Accepted: 02/22/2017] [Indexed: 06/07/2023]
Abstract
The purpose of this study was to investigate whether low-intensity pulsed ultrasound (LIPUS) promotes exercise-induced muscle hypertrophy. Twenty-four adult Sprague-Dawley (SD) rats were randomly assigned to three groups (n = 8 per group): normal control group (NC), treadmill exercise group (TE) and treadmill exercise + LIPUS group (TE + LIPUS). The TE + LIPUS group received a LIPUS treatment (1 MHz, 30 mW/cm2) at the gastrocnemius for 20 min/d after treadmill exercise. The TE group was sham-treated. Eight weeks of treadmill training successfully established the exercise-induced muscle hypertrophy model. Muscle strength, muscle mass and muscle fiber cross-sectional area were significantly increased in the TE + LIPUS group compared with the TE group. Moreover, LIPUS treatment significantly upregulated the expression of Akt, mTOR, p-Akt and p-mTOR and significantly downregulated the expression of MSTN, ActRIIB, FoxO1 and its phosphorylation. The results indicated that LIPUS promotes exercise-induced muscle hypertrophy by facilitating protein synthesis and inhibiting the protein catabolism pathway.
Collapse
Affiliation(s)
- Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Jing Zhang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Xinjuan Zhao
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Nan Li
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Wenqi Jian
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Shuxin Sun
- Department of Electronic Engineering, Fudan University, Shanghai, China
| | - Jianzhong Guo
- Shaanxi Key Laboratory of Ultrasonics, Shaanxi Normal University, Xi'an, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China.
| | - Dean Ta
- Department of Electronic Engineering, Fudan University, Shanghai, China; State Key Laboratory of ASIC and System, Fudan University, Shanghai, China; Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai, China
| |
Collapse
|
42
|
He L, Huang N, Li H, Tian J, Zhou X, Li T, Yao K, Wu G, Yin Y. AMPK/α-Ketoglutarate Axis Regulates Intestinal Water and Ion Homeostasis in Young Pigs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:2287-2298. [PMID: 28241728 DOI: 10.1021/acs.jafc.7b00324] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Water and ion absorption via sensitive aquaporins (AQPs) and ion channels is of critical importance in intestinal health. However, whether α-ketoglutarate (AKG) could improve intestinal water and ion homeostasis in lipopolysaccharide (LPS)-challenged piglets and whether the AMP-activated protein kinase (AMPK) pathway is involved remains largely unknown. This study was conducted to investigate the effect of dietary AKG supplementation on the small intestinal water and ion homeostasis through modulating the AMPK pathway in a piglet diarrhea model. A total of 32 weaned piglets were used in a 2 × 2 factorial design; the major factors were diet (basal diet or 1% AKG diet) and challenge (Escherichia coli LPS or saline). The results showed that LPS challenge increased the diarrhea index and affected the concentrations of serum Na+, K+, Cl-, glucose, and AKG and its metabolites in piglets fed the basal or AKG diet. However, the addition of AKG attenuated diarrhea incidence and reversed these serum parameter concentrations. Most AQPs (e.g., AQP1, AQP3, AQP4, AQP5, AQP8, AQP10, and AQP11) and ion transporters (NHE3, ENaC, and DRA/PAT1) were widely distributed in the duodenum and jejunum of piglets. We also found that AKG up-regulated the expression of intestinal epithelial AQPs while inhibiting the expression of ion transporters. LPS challenge decreased (P < 0.05) the gene and protein expression of the AMPK pathway (AMPKα1, AMPKα2, SIRT1, PGC-1α, ACC, and TORC2) in the jejunum and ileum. Notably, AKG supplementation enhanced the abundance of these proteins in the LPS-challenged piglets. Collectively, AKG plays an important role in increasing water and ion homeostasis through modulating the AMPK pathway. Our novel finding has important implications for the prevention and treatment of gut dysfunction in neonates.
Collapse
Affiliation(s)
- Liuqin He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production , Changsha, Hunan 410125, China
- University of the Chinese Academy of Sciences , Beijing 10008, China
| | - Niu Huang
- College of Animal Science and Technology, Hunan Agricultural University , Changsha, Hunan 410128, China
| | - Huan Li
- College of Animal Science and Technology, Hunan Agricultural University , Changsha, Hunan 410128, China
| | - Junquan Tian
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production , Changsha, Hunan 410125, China
- University of the Chinese Academy of Sciences , Beijing 10008, China
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production , Changsha, Hunan 410125, China
| | - Tiejun Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production , Changsha, Hunan 410125, China
| | - Kang Yao
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production , Changsha, Hunan 410125, China
- College of Animal Science and Technology, Hunan Agricultural University , Changsha, Hunan 410128, China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University , College Station, Texas 77843, United States
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production , Changsha, Hunan 410125, China
| |
Collapse
|
43
|
Wu J, Zhu C, Yang L, Wang Z, Wang L, Wang S, Gao P, Zhang Y, Jiang Q, Zhu X, Shu G. N-Oleoylglycine-Induced Hyperphagia Is Associated with the Activation of Agouti-Related Protein (AgRP) Neuron by Cannabinoid Receptor Type 1 (CB1R). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:1051-1057. [PMID: 28102080 DOI: 10.1021/acs.jafc.6b05281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
N-Acyl amino acids (NAAAs) are conjugate products of fatty acids and amino acids, which are available in animal-derived food. We compared the effects of N-arachidonoylglycine (NAGly), N-arachidonoylserine (NASer), and N-oleoylglycine (OLGly) on in vivo food intake and in vitro [Ca2+]i of Agouti-related protein (AgRP) neurons to identify the role of these compounds in energy homeostasis. Hypothalamic neuropeptide expression and anxiety behavior in response to OLGly were also tested. To further identify the underlying mechanism of OLGly on food intake, we first detected the expression level of potential OLGly receptors. The cannabinoid receptor type 1 (CB1R) antagonist was cotreated with OLGly to analyze the activation of AgRP neuron, including [Ca2+]i, expression levels of PKA, CREB, and c-Fos, and neuropeptide secretion. Results demonstrated that only OLGly (intrapertioneal injection of 6 mg/kg) can induce hyperphagia without changing the expression of hypothalamic neuropeptides and anxiety-like behavior. Moreover, 20 μM OLGly robustly enhances [Ca2+]i, c-Fos protein expression in AgRP neuron, and AgRP content in the culture medium. OLGly-induced activation of AgRP neuron was completely abolished by the CB1R-specific antagonist, AM251. In summary, this study is the first to demonstrate the association of OLGly-induced hyperphagia with activation of the AgRP neuron by CB1R. These findings open avenues for investigation and application of OLGly to modulate energy homeostasis.
Collapse
Affiliation(s)
- Junguo Wu
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Canjun Zhu
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Liusong Yang
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Zhonggang Wang
- Huahong Engineering and Research Center of Agricultural and Livestock , Zhaoqing, Guangdong 430051, People's Republic of China
| | - Lina Wang
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Songbo Wang
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Ping Gao
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Yongliang Zhang
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Qingyan Jiang
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Xiaotong Zhu
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| | - Gang Shu
- College of Animal Science & National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510640, People's Republic of China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, People's Republic of China
| |
Collapse
|
44
|
Meng Y, Zhang J, Zhang F, Ai W, Zhu X, Shu G, Wang L, Gao P, Xi Q, Zhang Y, Liang X, Jiang Q, Wang S. Lauric Acid Stimulates Mammary Gland Development of Pubertal Mice through Activation of GPR84 and PI3K/Akt Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:95-103. [PMID: 27978622 DOI: 10.1021/acs.jafc.6b04878] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
It has been demonstrated that dietary fat affects pubertal mammary gland development. However, the role of lauric acid (LA) in this process remains unclear. Thus, this study aimed to investigate the effects of LA on mammary gland development in pubertal mice and to explore the underlying mechanism. In vitro, 100 μM LA significantly promoted proliferation of mouse mammary epithelial cell line HC11 by regulating expression of proliferative markers (cyclin D1/3, p21, PCNA). Meanwhile, LA activated the G protein-coupled receptor 84 (GPR84) and PI3K/Akt signaling pathway. In agreement, dietary 1% LA enhanced mammary duct development, increased the expression of GPR84 and cyclin D1, and activated PI3K/Akt in mammary gland of pubertal mice. Furthermore, knockdown of GPR84 or inhibition of PI3K/Akt totally abolished the promotion of HC11 proliferation induced by LA. These results showed that LA stimulated mammary gland development of pubertal mice through activation of GPR84 and PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yingying Meng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Jing Zhang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Fenglin Zhang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Wei Ai
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Xiaotong Zhu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Gang Shu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Lina Wang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Ping Gao
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Qianyun Xi
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Yongliang Zhang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Xingwei Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi High Education Laboratory for Animal Reproduction and Biotechnology, Guangxi University , Nanning 530004, P. R. China
| | - Qingyan Jiang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| | - Songbo Wang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University , Guangzhou 510642, P. R. China
- ALLTECH-SCAU Animal Nutrition Control Research Alliance, South China Agricultural University , Guangzhou 510642, P. R. China
| |
Collapse
|
45
|
TSC2 Deficiency Unmasks a Novel Necrosis Pathway That Is Suppressed by the RIP1/RIP3/MLKL Signaling Cascade. Cancer Res 2016; 76:7130-7139. [DOI: 10.1158/0008-5472.can-16-1052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/09/2016] [Accepted: 10/07/2016] [Indexed: 11/16/2022]
|