1
|
Sunilkumar S, Dennis MD. REDD1 Is a Promising Therapeutic Target to Combat the Development of Diabetes Complications: A Report on Research Supported by Pathway to Stop Diabetes. Diabetes 2024; 73:1553-1562. [PMID: 38976480 PMCID: PMC11417436 DOI: 10.2337/dbi24-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
The stress response protein regulated in development and DNA damage response 1 (REDD1) has emerged as a key player in the pathogenesis of diabetes. Diabetes upregulates REDD1 in a variety of insulin-sensitive tissues, where the protein acts to inhibit signal transduction downstream of the insulin receptor. REDD1 functions as a cytosolic redox sensor that suppresses Akt/mTORC1 signaling to reduce energy expenditure in response to cellular stress. Whereas a transient increase in REDD1 contributes to an adaptive cellular response, chronically elevated REDD1 levels are implicated in disease progression. Recent studies highlight the remarkable benefits of both whole-body and tissue-specific REDD1 deletion in preclinical models of type 1 and type 2 diabetes. In particular, REDD1 is necessary for the development of glucose intolerance and the consequent rise in oxidative stress and inflammation. Here, we review studies that support a role for chronically elevated REDD1 levels in the development of diabetes complications, reflect on limitations of prior therapeutic approaches targeting REDD1 in patients, and discuss potential opportunities for future interventions to improve the lives of people living with diabetes. This article is part of a series of Perspectives that report on research funded by the American Diabetes Association Pathway to Stop Diabetes program. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Siddharth Sunilkumar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Michael D. Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| |
Collapse
|
2
|
Kato H, Salgado M, Mendez D, Gonzalez N, Rawson J, Ligot D, Balandran B, Orr C, Quijano JC, Omori K, Qi M, Al-Abdullah IH, Mullen Y, Ku HT, Kandeel F, Komatsu H. Biological hypoxia in pre-transplant human pancreatic islets induces transplant failure in diabetic mice. Sci Rep 2024; 14:12402. [PMID: 38811610 PMCID: PMC11137081 DOI: 10.1038/s41598-024-61604-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Evaluating the quality of isolated human islets before transplantation is crucial for predicting the success in treating Type 1 diabetes. The current gold standard involves time-intensive in vivo transplantation into diabetic immunodeficient mice. Given the susceptibility of isolated islets to hypoxia, we hypothesized that hypoxia present in islets before transplantation could indicate compromised islet quality, potentially leading to unfavorable outcomes. To test this hypothesis, we analyzed expression of 39 hypoxia-related genes in human islets from 85 deceased donors. We correlated gene expression profiles with transplantation outcomes in 327 diabetic mice, each receiving 1200 islet equivalents grafted into the kidney capsule. Transplantation outcome was post-transplant glycemic control based on area under the curve of blood glucose over 4 weeks. In linear regression analysis, DDIT4 (R = 0.4971, P < 0.0001), SLC2A8 (R = 0.3531, P = 0.0009) and HK1 (R = 0.3444, P = 0.0012) had the highest correlation with transplantation outcome. A multiple regression model of 11 genes increased the correlation (R = 0.6117, P < 0.0001). We conclude that assessing pre-transplant hypoxia in human islets via gene expression analysis is a rapid, viable alternative to conventional in vivo assessments. This approach also underscores the importance of mitigating pre-transplant hypoxia in isolated islets to improve the success rate of islet transplantation.
Collapse
Affiliation(s)
- Hiroyuki Kato
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
- Department of Surgery, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA
| | - Mayra Salgado
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Daniel Mendez
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Nelson Gonzalez
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Jeffrey Rawson
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Doreen Ligot
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Bennie Balandran
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Chris Orr
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Janine C Quijano
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Keiko Omori
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Meirigeng Qi
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Ismail H Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Yoko Mullen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Hsun Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Hirotake Komatsu
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA.
- Department of Surgery, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA.
| |
Collapse
|
3
|
Couvert A, Lacaze L, Touboulic S, Gautier S, Guérin S, Randuineau G, Romé V, Malbert CH, Val-Laillet D, Derbré F, Thibault R. The Yucatan minipig model: A new preclinical model of malnutrition in obese patients with acute or chronic diseases. Clin Nutr 2024; 43:357-365. [PMID: 38142480 DOI: 10.1016/j.clnu.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND & AIMS Malnutrition can develop in patients with obesity suffering from acute or chronic illness or after obesity surgery, promoting sarcopenic obesity. A better understanding of this pathophysiology and the development of new therapeutics for chronic diseases, that are often complicated with malnutrition and obesity, justify the development of new animal experimental models close to the human physiology. This study aims to characterize the effects of obesity and underfeeding on Yucatan obese minipigs, assessing its validity as a preclinical model for obesity-related malnutrition. METHODS Sixteen 30-month-old Yucatan minipigs were divided into two groups for 8 weeks: a standard diet group (ST, n = 5) and an obesogenic diet group (OB, n = 11). After 8 weeks, the OB group was further divided into two sub-groups: a standard diet group (OB-ST, n = 5) and a low-calorie/low-protein diet group (OB-LC/LP, n = 6) for 8 weeks. Body composition by CT-Scan and blood parameters were monitored, and trapezius muscle biopsies were collected to analyse signaling pathways involved in protein turnover and energy metabolism. RESULTS At W8, OB-ST animals exhibited significantly higher body weight (+37.7%, p = 0.03), muscle mass (+24.9%, p = 0.02), and visceral fat (+192.0%, p = 0.03) compared to ST. Trapezius cross sectional area (CSA) normalized to body weight was lower in OB-ST animals (-15.02%, p = 0.017). At W16, no significant changes were observed in protein turnover markers, although REDD1 increased in OB-ST (96.4%, p = 0.02). After 8 weeks of low-caloric/low protein diet, OB-LC/LP showed decreased body weight (-9.8%, p = 0.03), muscle mass (-6.5%, p = 0.03), and visceral fat (-41.5%, p = 0.03) compared to OB-ST animals. Trapezius fiber CSA significantly decreased in OB-LC/LP (-36.1%, p < 0.0001) and normalized to body weight (-25.4%, p < 0.0001), combined to higher ubiquitinated protein content (+38.3%, p = 0.02). CONCLUSION Our data support that the Yucatan minipig model mimics nutritional and skeletal muscle phenotypes observed in obese patients, with or without protein-energy malnutrition. It also reproduces muscle atrophy observed in chronic diseases or post-obesity surgery, making it a promising preclinical model for obesity-related malnutrition.
Collapse
Affiliation(s)
- Annaëlle Couvert
- Laboratory "Movement Sport and Health Sciences" EA 7470, University of Rennes, ENS Rennes, 35170 Bruz, France; Service Endocrinologie-Diabétologie-Nutrition, Centre labellisé de nutrition parentérale au domicile, CHU Rennes, Rennes, France
| | - Laurence Lacaze
- Service Endocrinologie-Diabétologie-Nutrition, Centre labellisé de nutrition parentérale au domicile, CHU Rennes, Rennes, France; INRAE, INSERM, Univ Rennes, NuMeCan, Nutrition Metabolisms Cancer, Rennes, France
| | - Steve Touboulic
- INRAE, INSERM, Univ Rennes, NuMeCan, Nutrition Metabolisms Cancer, Rennes, France
| | - Sandrine Gautier
- Laboratory "Movement Sport and Health Sciences" EA 7470, University of Rennes, ENS Rennes, 35170 Bruz, France
| | - Sylvie Guérin
- INRAE, INSERM, Univ Rennes, NuMeCan, Nutrition Metabolisms Cancer, Rennes, France
| | - Gwénaëlle Randuineau
- INRAE, INSERM, Univ Rennes, NuMeCan, Nutrition Metabolisms Cancer, Rennes, France
| | - Véronique Romé
- INRAE, INSERM, Univ Rennes, NuMeCan, Nutrition Metabolisms Cancer, Rennes, France
| | | | - David Val-Laillet
- INRAE, INSERM, Univ Rennes, NuMeCan, Nutrition Metabolisms Cancer, Rennes, France
| | - Frédéric Derbré
- Laboratory "Movement Sport and Health Sciences" EA 7470, University of Rennes, ENS Rennes, 35170 Bruz, France.
| | - Ronan Thibault
- Service Endocrinologie-Diabétologie-Nutrition, Centre labellisé de nutrition parentérale au domicile, CHU Rennes, Rennes, France; INRAE, INSERM, Univ Rennes, NuMeCan, Nutrition Metabolisms Cancer, Rennes, France.
| |
Collapse
|
4
|
Kim JY, Kwon YG, Kim YM. The stress-responsive protein REDD1 and its pathophysiological functions. Exp Mol Med 2023; 55:1933-1944. [PMID: 37653030 PMCID: PMC10545776 DOI: 10.1038/s12276-023-01056-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/13/2023] [Accepted: 05/17/2023] [Indexed: 09/02/2023] Open
Abstract
Regulated in development and DNA damage-response 1 (REDD1) is a stress-induced protein that controls various cellular functions, including metabolism, oxidative stress, autophagy, and cell fate, and contributes to the pathogenesis of metabolic and inflammatory disorders, neurodegeneration, and cancer. REDD1 usually exerts deleterious effects, including tumorigenesis, metabolic inflammation, neurodegeneration, and muscle dystrophy; however, it also exhibits protective functions by regulating multiple intrinsic cell activities through either an mTORC1-dependent or -independent mechanism. REDD1 typically regulates mTORC1 signaling, NF-κB activation, and cellular pro-oxidant or antioxidant activity by interacting with 14-3-3 proteins, IκBα, and thioredoxin-interacting protein or 75 kDa glucose-regulated protein, respectively. The diverse functions of REDD1 depend on cell type, cellular context, interaction partners, and cellular localization (e.g., mitochondria, endomembrane, or cytosol). Therefore, comprehensively understanding the molecular mechanisms and biological roles of REDD1 under pathophysiological conditions is of utmost importance. In this review, based on the published literature, we highlight and discuss the molecular mechanisms underlying the REDD1 expression and its actions, biological functions, and pathophysiological roles.
Collapse
Affiliation(s)
- Ji-Yoon Kim
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul, 04763, Republic of Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
5
|
Stevens SA, Gonzalez Aguiar MK, Toro AL, Yerlikaya EI, Sunilkumar S, VanCleave AM, Pfleger J, Bradley EA, Kimball SR, Dennis MD. PERK/ATF4-dependent expression of the stress response protein REDD1 promotes proinflammatory cytokine expression in the heart of obese mice. Am J Physiol Endocrinol Metab 2023; 324:E62-E72. [PMID: 36383638 PMCID: PMC9870577 DOI: 10.1152/ajpendo.00238.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Endoplasmic reticulum (ER) stress and inflammation are hallmarks of myocardial impairment. Here, we investigated the role of the stress response protein regulated in development and DNA damage 1 (REDD1) as a molecular link between ER stress and inflammation in cardiomyocytes. In mice fed a high-fat high-sucrose (HFHS, 42% kcal fat, 34% sucrose by weight) diet for 12 wk, REDD1 expression in the heart was increased in coordination with markers of ER stress and inflammation. In human AC16 cardiomyocytes exposed to either hyperglycemic conditions or the saturated fatty acid palmitate, REDD1 expression was increased coincident with ER stress and upregulated expression of the proinflammatory cytokines IL-1β, IL-6, and TNFα. In cardiomyocytes exposed to hyperglycemic/hyperlipidemic conditions, pharmacological inhibition of the ER kinase protein kinase RNA-like endoplasmic reticulum kinase (PERK) or knockdown of the transcription factor ATF4 prevented the increase in REDD1 expression. REDD1 deletion reduced proinflammatory cytokine expression in both cardiomyocytes exposed to hyperglycemic/hyperlipidemic conditions and in the hearts of obese mice. Overall, the findings support a model wherein HFHS diet contributes to the development of inflammation in cardiomyocytes by promoting REDD1 expression via activation of a PERK/ATF4 signaling axis.NEW & NOTEWORTHY Interplay between endoplasmic reticulum stress and inflammation contributes to cardiovascular disease progression. The studies here identify the stress response protein known as REDD1 as a missing molecular link that connects the development of endoplasmic reticulum stress with increased production of proinflammatory cytokines in the hearts of obese mice.
Collapse
Affiliation(s)
- Shaunaci A Stevens
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Maria K Gonzalez Aguiar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Allyson L Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Esma I Yerlikaya
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Siddharth Sunilkumar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Ashley M VanCleave
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Jessica Pfleger
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia
| | - Elisa A Bradley
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
- Division of Cardiovascular Medicine, Penn State Health Heart and Vascular Institute, Hershey S. Milton Medical Center, Hershey, Pennsylvania
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Michael D Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
6
|
Suryawan A, Rudar M, Naberhuis JK, Fiorotto ML, Davis TA. Preterm birth alters the feeding-induced activation of Akt signaling in the muscle of neonatal piglets. Pediatr Res 2022:10.1038/s41390-022-02382-4. [PMID: 36402914 DOI: 10.1038/s41390-022-02382-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/04/2022] [Accepted: 10/27/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Postnatal lean mass accretion is commonly reduced in preterm infants. This study investigated mechanisms involved in the blunted feeding-induced activation of Akt in the skeletal muscle of preterm pigs that contributes to lower protein synthesis rates. METHODS On day 3 following cesarean section, preterm and term piglets were fasted or fed an enteral meal. Activation of Akt signaling pathways in skeletal muscle was determined. RESULTS Akt1 and Akt2, but not Akt3, phosphorylation were lower in the skeletal muscle of preterm than in term pigs (P < 0.05). Activation of Akt-positive regulators, PDK1 and mTORC2, but not FAK, were lower in preterm than in term (P < 0.05). The formation of Akt complexes with GAPDH and Hsp90 and the abundance of Ubl4A were lower in preterm than in term (P < 0.05). The abundance of Akt inhibitors, PHLPP and SHIP2, but not PTEN and IP6K1, were higher in preterm than in term pigs (P < 0.05). PP2A activation was inhibited by feeding in term but not in preterm pigs (P < 0.05). CONCLUSIONS Our results suggest that preterm birth impairs regulatory components involved in Akt activation, thereby limiting the anabolic response to feeding. This anabolic resistance likely contributes to the reduced lean accretion following preterm birth. IMPACT The Akt-mTORC1 pathway plays an important role in the regulation of skeletal muscle protein synthesis in neonates. This is the first evidence to demonstrate that, following preterm birth, the postprandial activation of positive regulators of Akt in the skeletal muscle is reduced, whereas the activation of negative regulators of Akt is enhanced. This anabolic resistance of Akt signaling in response to feeding likely contributes to the reduced accretion of lean mass in premature infants. These results may provide potential novel molecular targets for intervention to enhance lean growth in preterm neonates.
Collapse
Affiliation(s)
- Agus Suryawan
- United States Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Marko Rudar
- Department of Animal Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Jane K Naberhuis
- United States Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Marta L Fiorotto
- United States Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Teresa A Davis
- United States Department of Agriculture/Agricultural Research Service, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Sunilkumar S, Yerlikaya EI, Toro AL, Miller WP, Chen H, Hu K, Kimball SR, Dennis MD. REDD1 Ablation Attenuates the Development of Renal Complications in Diabetic Mice. Diabetes 2022; 71:2412-2425. [PMID: 35984399 PMCID: PMC9630083 DOI: 10.2337/db22-0402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/17/2022] [Indexed: 02/03/2023]
Abstract
Chronic hyperglycemia contributes to development of diabetic kidney disease by promoting glomerular injury. In this study, we evaluated the hypothesis that hyperglycemic conditions promote expression of the stress response protein regulated in development and DNA damage response 1 (REDD1) in the kidney in a manner that contributes to the development of oxidative stress and renal injury. After 16 weeks of streptozotocin-induced diabetes, albuminuria and renal hypertrophy were observed in wild-type (WT) mice coincident with increased renal REDD1 expression. In contrast, diabetic REDD1 knockout (KO) mice did not exhibit impaired renal physiology. Histopathologic examination revealed that glomerular damage including mesangial expansion, matrix deposition, and podocytopenia in the kidneys of diabetic WT mice was reduced or absent in diabetic REDD1 KO mice. In cultured human podocytes, exposure to hyperglycemic conditions enhanced REDD1 expression, increased reactive oxygen species (ROS) levels, and promoted cell death. In both the kidney of diabetic mice and in podocyte cultures exposed to hyperglycemic conditions, REDD1 deletion reduced ROS and prevented podocyte loss. Benefits of REDD1 deletion were recapitulated by pharmacological GSK3β suppression, supporting a role for REDD1-dependent GSK3β activation in diabetes-induced oxidative stress and renal defects. The results support a role for REDD1 in diabetes-induced renal complications.
Collapse
Affiliation(s)
- Siddharth Sunilkumar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Esma I. Yerlikaya
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Allyson L. Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - William P. Miller
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Han Chen
- Transmission Electron Microscopy Core, Penn State College of Medicine, Hershey, PA
| | - Kebin Hu
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
- Division of Nephrology, Department of Medicine, Penn State College of Medicine, Hershey, PA
| | - Scot R. Kimball
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Michael D. Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
- Corresponding author: Michael D. Dennis,
| |
Collapse
|
8
|
Nutritional Sensor REDD1 in Cancer and Inflammation: Friend or Foe? Int J Mol Sci 2022; 23:ijms23179686. [PMID: 36077083 PMCID: PMC9456073 DOI: 10.3390/ijms23179686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
Regulated in Development and DNA Damage Response 1 (REDD1)/DNA Damage-Induced Transcript 4 (DDIT4) is an immediate early response gene activated by different stress conditions, including growth factor depletion, hypoxia, DNA damage, and stress hormones, i.e., glucocorticoids. The most known functions of REDD1 are the inhibition of proliferative signaling and the regulation of metabolism via the repression of the central regulator of these processes, the mammalian target of rapamycin (mTOR). The involvement of REDD1 in cell growth, apoptosis, metabolism, and oxidative stress implies its role in various pathological conditions, including cancer and inflammatory diseases. Recently, REDD1 was identified as one of the central genes mechanistically involved in undesirable atrophic effects induced by chronic topical and systemic glucocorticoids widely used for the treatment of blood cancer and inflammatory diseases. In this review, we discuss the role of REDD1 in the regulation of cell signaling and processes in normal and cancer cells, its involvement in the pathogenesis of different diseases, and the approach to safer glucocorticoid receptor (GR)-targeted therapies via a combination of glucocorticoids and REDD1 inhibitors to decrease the adverse atrophogenic effects of these steroids.
Collapse
|
9
|
Chaves AB, Zheng D, Johnson JA, Bergman BC, Patinkin ZW, Zaegel V, Biagioni EM, Krassovskaia P, Broskey NT, May LE, Dabelea D, Houmard JA, Boyle KE. Infant Mesenchymal Stem Cell Insulin Action Is Associated With Maternal Plasma Free Fatty Acids, Independent of Obesity Status: The Healthy Start Study. Diabetes 2022; 71:1649-1659. [PMID: 35621990 PMCID: PMC9490356 DOI: 10.2337/db21-0812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 05/16/2022] [Indexed: 11/13/2022]
Abstract
Preclinical rodent and nonhuman primate models investigating maternal obesity have highlighted the importance of the intrauterine environment in the development of insulin resistance in offspring; however, it remains unclear if these findings can be translated to humans. To investigate possible intrauterine effects in humans, we isolated mesenchymal stem cells (MSCs) from the umbilical cord tissue of infants born to mothers of normal weight or mothers with obesity. Insulin-stimulated glycogen storage was determined in MSCs undergoing myogenesis in vitro. There was no difference in insulin action based on maternal obesity. However, maternal free fatty acid (FFA) concentration, cord leptin, and intracellular triglyceride content were positively correlated with insulin action. Furthermore, MSCs from offspring born to mothers with elevated FFAs displayed elevated activation of the mTOR signaling pathway. Taken together, these data suggest that infants born to mothers with elevated lipid availability have greater insulin action in MSCs, which may indicate upregulation of growth and lipid storage pathways during periods of maternal overnutrition.
Collapse
Affiliation(s)
- Alec B. Chaves
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Donghai Zheng
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Jonathan A. Johnson
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Bryan C. Bergman
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Zachary W. Patinkin
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY
| | - Vincent Zaegel
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Ericka M. Biagioni
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Polina Krassovskaia
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Nicholas T. Broskey
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Linda E. May
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Dana Dabelea
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO
- The Lifecourse Epidemiology of Adiposity and Diabetes Center, Aurora, CO
| | - Joseph A. Houmard
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Kristen E. Boyle
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- The Lifecourse Epidemiology of Adiposity and Diabetes Center, Aurora, CO
| |
Collapse
|
10
|
Park M, Kim J, Kim T, Kim S, Park W, Ha KS, Cho SH, Won MH, Lee JH, Kwon YG, Kim YM. REDD1 is a determinant of low-dose metronomic doxorubicin-elicited endothelial cell dysfunction through downregulation of VEGFR-2/3 expression. Exp Mol Med 2021; 53:1612-1622. [PMID: 34697389 PMCID: PMC8568908 DOI: 10.1038/s12276-021-00690-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/21/2021] [Accepted: 06/29/2021] [Indexed: 01/10/2023] Open
Abstract
Low-dose metronomic chemotherapy (LDMC) inhibits tumor angiogenesis and growth by targeting tumor-associated endothelial cells, but the molecular mechanism has not been fully elucidated. Here, we examined the functional role of regulated in development and DNA damage responses 1 (REDD1), an inhibitor of mammalian target of rapamycin complex 1 (mTORC1), in LDMC-mediated endothelial cell dysfunction. Low-dose doxorubicin (DOX) treatment induced REDD1 expression in cultured vascular and lymphatic endothelial cells and subsequently repressed the mRNA expression of mTORC1-dependent translation of vascular endothelial growth factor receptor (Vegfr)-2/3, resulting in the inhibition of VEGF-mediated angiogenesis and lymphangiogenesis. These regulatory effects of DOX-induced REDD1 expression were additionally confirmed by loss- and gain-of-function studies. Furthermore, LDMC with DOX significantly suppressed tumor angiogenesis, lymphangiogenesis, vascular permeability, growth, and metastasis in B16 melanoma-bearing wild-type but not Redd1-deficient mice. Altogether, our findings indicate that REDD1 is a crucial determinant of LDMC-mediated functional dysregulation of tumor vascular and lymphatic endothelial cells by translational repression of Vegfr-2/3 transcripts, supporting the potential therapeutic properties of REDD1 in highly progressive or metastatic tumors.
Collapse
Affiliation(s)
- Minsik Park
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Joohwan Kim
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Taesam Kim
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Suji Kim
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Wonjin Park
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Kwon-Soo Ha
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Sung Hwan Cho
- grid.412010.60000 0001 0707 9039Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Moo-Ho Won
- grid.412010.60000 0001 0707 9039Department of Neurobiology, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Jeong-Hyung Lee
- grid.412010.60000 0001 0707 9039Department of Biochemistry, Kangwon National University, Chuncheon, Gangwon-Do 24341 Republic of Korea
| | - Young-Guen Kwon
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Republic of Korea
| | - Young-Myeong Kim
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea ,grid.412010.60000 0001 0707 9039Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-do 24341 Republic of Korea
| |
Collapse
|
11
|
Hain BA, Xu H, VanCleave AM, Gordon BS, Kimball SR, Waning DL. REDD1 deletion attenuates cancer cachexia in mice. J Appl Physiol (1985) 2021; 131:1718-1730. [PMID: 34672766 PMCID: PMC10392697 DOI: 10.1152/japplphysiol.00536.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cancer cachexia is a wasting disorder associated with advanced cancer that contributes to mortality. Cachexia is characterized by involuntary loss of body weight and muscle weakness that affects physical function. Regulated in DNA damage and development 1 (REDD1) is a stress-response protein that is transcriptionally upregulated in muscle during wasting conditions and inhibits mechanistic target of rapamycin complex 1 (mTORC1). C2C12 myotubes treated with Lewis lung carcinoma (LLC)-conditioned media increased REDD1 mRNA expression and decreased myotube diameter. To investigate the role of REDD1 in cancer cachexia, we inoculated 12-week old male wild-type or global REDD1 knockout (REDD1 KO) mice with LLC cells and euthanized 28-days later. Wild-type mice had increased skeletal muscle REDD1 expression, and REDD1 deletion prevented loss of body weight and lean tissue mass, but not fat mass. We found that REDD1 deletion attenuated loss of individual muscle weights and loss of myofiber cross sectional area. We measured markers of the Akt/mTORC1 pathway and found that, unlike wild-type mice, phosphorylation of both Akt and 4E-BP1 was maintained in the muscle of REDD1 KO mice after LLC inoculation, suggesting that loss of REDD1 is beneficial in maintaining mTORC1 activity in mice with cancer cachexia. We measured Foxo3a phosphorylation as a marker of the ubiquitin proteasome pathway and autophagy and found that REDD1 deletion prevented dephosphorylation of Foxo3a in muscles from cachectic mice. Our data provides evidence that REDD1 plays an important role in cancer cachexia through the regulation of both protein synthesis and protein degradation pathways.
Collapse
Affiliation(s)
- Brian A Hain
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States.,Penn State Cancer Institute, Hershey, PA, United States
| | - Haifang Xu
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States
| | - Ashley M VanCleave
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States
| | - Bradley S Gordon
- Florida State University, Dept. of Nutrition and Integrative Physiology, Tallahassee, FL, United States
| | - Scot R Kimball
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States
| | - David L Waning
- The Penn State College of Medicine, Dept. of Cellular and Molecular Physiology, Hershey, PA, United States.,Penn State Cancer Institute, Hershey, PA, United States
| |
Collapse
|
12
|
Huang P, Bai L, Liu L, Fu J, Wu K, Liu H, Liu Y, Qi B, Qi B. Redd1 knockdown prevents doxorubicin-induced cardiac senescence. Aging (Albany NY) 2021; 13:13788-13806. [PMID: 33962393 PMCID: PMC8202877 DOI: 10.18632/aging.202972] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Regulated in development and DNA damage response-1 (Redd1) is a stress-response gene that is transcriptionally induced by diverse stressful stimuli to influence cellular growth and survival. Although evidence suggests that aging may drive Redd1 expression in skeletal muscles, the expression patterns and functions of Redd1 in senescent cardiomyocytes remain unspecified. To address this issue, in vitro and in vivo models of cardiomyocyte senescence were established by administration of doxorubicin (Dox). Redd1 overexpression and knockdown was achieved in cultured H9c2 cardiomyocytes and mouse tissues using, respectively, lentivirals and adeno-associated virus 9 (AAV9) vectors. In the hearts of both aged (24 months old) and Dox-treated mice, as well as in Dox-exposed H9c2 cardiomyocytes, high Redd1 expression accompanied the increase in both cellular senescence markers (p16INK4a and p21) and pro-inflammatory cytokine expression indicative of a stress-associated secretory phenotype (SASP). Notably, Redd1 overexpression accentuated, whereas Redd1 silencing markedly attenuated, Dox-induced cardiomyocyte senescence features both in vitro and in vivo. Notably, AAV9-shRNA-mediated Redd1 silencing significantly alleviated Dox-induced cardiac dysfunction. Moreover, through pharmacological inhibition, immunofluorescence, and western blotting, signaling pathway analyses indicated that Redd1 promotes cardiomyocyte senescence as a downstream effector of p38 MAPK to promote NF-kB signaling via p65 phosphorylation and nuclear translocation.
Collapse
Affiliation(s)
- Pianpian Huang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
- Department of Geriatrics, Wuhan No.1 Hospital, Wuhan, Hubei 430022, China
| | - Lijuan Bai
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Lihua Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jun Fu
- Department of Radiology, Wuhan No.1 Hospital, Wuhan, Hubei 430022, China
| | - Kefei Wu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hongxia Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Benming Qi
- Department of Otorhinolaryngology, First People’s Hospital of Yunnan Province, Kunming, Yunnan 650000, China
| | - Benling Qi
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
13
|
Britto FA, Dumas K, Giorgetti-Peraldi S, Ollendorff V, Favier FB. Is REDD1 a metabolic double agent? Lessons from physiology and pathology. Am J Physiol Cell Physiol 2020; 319:C807-C824. [PMID: 32877205 DOI: 10.1152/ajpcell.00340.2020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Akt/mechanistic target of rapamycin (mTOR) signaling pathway governs macromolecule synthesis, cell growth, and metabolism in response to nutrients and growth factors. Regulated in development and DNA damage response (REDD)1 is a conserved and ubiquitous protein, which is transiently induced in response to multiple stimuli. Acting like an endogenous inhibitor of the Akt/mTOR signaling pathway, REDD1 protein has been shown to regulate cell growth, mitochondrial function, oxidative stress, and apoptosis. Recent studies also indicate that timely REDD1 expression limits Akt/mTOR-dependent synthesis processes to spare energy during metabolic stresses, avoiding energy collapse and detrimental consequences. In contrast to this beneficial role for metabolic adaptation, REDD1 chronic expression appears involved in the pathogenesis of several diseases. Indeed, REDD1 expression is found as an early biomarker in many pathologies including inflammatory diseases, cancer, neurodegenerative disorders, depression, diabetes, and obesity. Moreover, prolonged REDD1 expression is associated with cell apoptosis, excessive reactive oxygen species (ROS) production, and inflammation activation leading to tissue damage. In this review, we decipher several mechanisms that make REDD1 a likely metabolic double agent depending on its duration of expression in different physiological and pathological contexts. We also discuss the role played by REDD1 in the cross talk between the Akt/mTOR signaling pathway and the energetic metabolism.
Collapse
Affiliation(s)
| | - Karine Dumas
- Université Cote d'Azur, INSERM, UMR1065, C3M, Nice, France
| | | | | | | |
Collapse
|
14
|
Rivera-Gonzalez GC, Klopot A, Sabin K, Baida G, Horsley V, Budunova I. Regulated in Development and DNA Damage Responses 1 Prevents Dermal Adipocyte Differentiation and Is Required for Hair Cycle-Dependent Dermal Adipose Expansion. J Invest Dermatol 2020; 140:1698-1705.e1. [PMID: 32032578 PMCID: PMC7398827 DOI: 10.1016/j.jid.2019.12.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/14/2019] [Accepted: 12/04/2019] [Indexed: 02/07/2023]
Abstract
Dermal white adipose tissue (dWAT) expansion is associated with important homeostatic and pathologic processes in skin. Even though mTOR/protein kinase B signaling is important for adipogenesis, the role of regulated development of DNA damage responses 1 (REDD1), a negative regulator of mTOR/protein kinase B, is poorly understood. Loss of REDD1 in mice resulted in reduction of body mass, total fat, size of gonadal white adipose tissue, and interscapular brown adipose tissue. Inguinal subcutaneous white adipose tissue and dWAT in REDD1 knockouts were expanded compared with wild type mice. Size and number of mature adipocytes in dWAT were also increased in adult REDD1 knockouts. This dWAT phenotype was established around postnatal day 18 and did not depend on the hair growth cycle. Numbers of adipocyte precursor cells were lower in REDD1 knockout skin. In vitro analysis revealed increased differentiation of skin-derived REDD1 knockout adipocyte precursor cells as indicated by higher lipid accumulation and increased adipogenic marker expression. 3T3L1 cells overexpressing REDD1 had decreased sensitivity to differentiation. Overall, our findings indicate that REDD1 silencing induced expansion of dWAT through hypertrophy and hyperplasia. This REDD1-dependent mechanism of adipogenesis could be used to preferentially target skin-associated adipose tissue for therapeutic purposes.
Collapse
Affiliation(s)
- Guillermo C. Rivera-Gonzalez
- Department of Molecular, Cellular and Developmental Biology and Department of Dermatology, Yale University, New Haven, CT 06520
- Current address: Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110
| | - Anna Klopot
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| | - Kaitlyn Sabin
- Department of Molecular, Cellular and Developmental Biology and Department of Dermatology, Yale University, New Haven, CT 06520
| | - Gleb Baida
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology and Department of Dermatology, Yale University, New Haven, CT 06520
| | - Irina Budunova
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| |
Collapse
|
15
|
Dumas K, Ayachi C, Gilleron J, Lacas‐Gervais S, Pastor F, Favier FB, Peraldi P, Vaillant N, Yvan‐Charvet L, Bonnafous S, Patouraux S, Anty R, Tran A, Gual P, Cormont M, Tanti J, Giorgetti‐Peraldi S. REDD1 deficiency protects against nonalcoholic hepatic steatosis induced by high‐fat diet. FASEB J 2020; 34:5046-5060. [DOI: 10.1096/fj.201901799rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Karine Dumas
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | - Chaima Ayachi
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | - Jerome Gilleron
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | | | - Faustine Pastor
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | | | - Pascal Peraldi
- Université Côte d’Azur, Inserm, CNRS, iBV, Team “Stem Cells and Differentiation” France
| | - Nathalie Vaillant
- Université Côte d’Azur, Inserm, C3M, Team “Haematometabolism in Diseases” France
| | - Laurent Yvan‐Charvet
- Université Côte d’Azur, Inserm, C3M, Team “Haematometabolism in Diseases” France
| | - Stéphanie Bonnafous
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
- Université Côte d’Azur, CHU, Inserm, C3M,Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Stéphanie Patouraux
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
- Université Côte d’Azur, CHU, Inserm, C3M,Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Rodolphe Anty
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
- Université Côte d’Azur, CHU, Inserm, C3M,Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Albert Tran
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
- Université Côte d’Azur, CHU, Inserm, C3M,Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Philippe Gual
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Mireille Cormont
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | - Jean‐François Tanti
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | - Sophie Giorgetti‐Peraldi
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| |
Collapse
|
16
|
Ato S, Kido K, Sato K, Fujita S. Type 2 diabetes causes skeletal muscle atrophy but does not impair resistance training-mediated myonuclear accretion and muscle mass gain in rats. Exp Physiol 2019; 104:1518-1531. [PMID: 31328833 PMCID: PMC6790689 DOI: 10.1113/ep087585] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
New Findings What is the central question of this study? Type 2 diabetes mellitus (T2DM) causes skeletal muscle atrophy; does it affect resistance training (RT)‐mediated molecular adaptations and subsequent muscle hypertrophy? What is the main finding and its importance? Although skeletal muscle mass and regulation were not preserved under conditions of T2DM, the response of RT‐induced skeletal muscle hypertrophy was not impaired in T2DM rat skeletal muscle. These findings suggest that the capacity of RT‐mediated muscle mass gain is not diminished in the T2DM condition.
Abstract Type 2 diabetes mellitus (T2DM) is known to cause skeletal muscle atrophy. However, it is not known whether T2DM affects resistance training (RT)‐mediated molecular adaptations and subsequent muscle hypertrophy. Therefore, we investigated the effect of T2DM on response of skeletal muscle hypertrophy to chronic RT using a rat resistance exercise mimetic model. T2DM and healthy control rats were subjected to 18 bouts (3 times per week) of chronic RT on unilateral lower legs. RT significantly increased gastrocnemius muscle mass and myonuclei in both T2DM and healthy control rats to the same extent, even though T2DM caused muscle atrophy in the resting condition. Further, T2DM significantly reduced mechanistic target of rapamycin complex 1 (mTORC1) activity (phosphorylation of p70S6KThr389 and 4E‐BP1Thr37/46) to insulin stimulation and the number of myonuclei in the untrained basal condition, but RT‐mediated adaptations were not affected by T2DM. These findings suggested that although the skeletal muscle mass and regulation were not preserved under basal conditions of T2DM, the response of RT‐induced skeletal muscle hypertrophy was not impaired in T2DM rat skeletal muscle.
Collapse
Affiliation(s)
- Satoru Ato
- Graduate School of Sport and Health Science, Ritsumeikan University, Kusatsu, Japan
| | - Kohei Kido
- Graduate School of Sport and Health Science, Ritsumeikan University, Kusatsu, Japan
| | - Koji Sato
- Faculty of Human Development, Kobe University, Kobe, Japan
| | - Satoshi Fujita
- Graduate School of Sport and Health Science, Ritsumeikan University, Kusatsu, Japan
| |
Collapse
|
17
|
Dungan CM, Gordon BS, Williamson DL. Acute treadmill exercise discriminately improves the skeletal muscle insulin-stimulated growth signaling responses in mice lacking REDD1. Physiol Rep 2019; 7:e14011. [PMID: 30806987 PMCID: PMC6383112 DOI: 10.14814/phy2.14011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
A loss of the regulated in development and DNA damage 1 (REDD1) hyperactivates mechanistic Target of Rapamycin Complex 1 (mTORC1) reducing insulin-stimulated insulin signaling, which could provide insight into mechanisms of insulin resistance. Although aerobic exercise acutely inhibits mTORC1 signaling, improvements in insulin-stimulated signaling are exhibited. The goal of this study was to determine if a single bout of treadmill exercise was sufficient to improve insulin signaling in mice lacking REDD1. REDD1 wildtype (WT) and REDD1 knockout (KO) mice were acutely exercised on a treadmill (30 min, 20 m/min, 5% grade). A within animal noninsulin-to-insulin-stimulated percent change in skeletal muscle insulin-stimulated kinases (IRS-1, ERK1/2, Akt), growth signaling activation (4E-BP1, S6K1), and markers of growth repression (REDD1, AMPK, FOXO1/3A) was examined, following no exercise control or an acute bout of exercise. Unlike REDD1 KO mice, REDD1 WT mice exhibited an increase (P < 0.05) in REDD1 following treadmill exercise. However, both REDD1 WT and KO mice exhibited an increase (P < 0.05) AMPK phosphorylation, and a subsequent reduction (P < 0.05) in mTORC1 signaling after the exercise bout versus nonexercising WT or KO mice. Exercise increased (P < 0.05) the noninsulin-to-insulin-stimulated percent change phosphorylation of mTORC1, ERK1/2, IRS-1, and Akt on S473 in REDD1 KO mice when compared to nonexercised KO mice. However, there was no change in the noninsulin-to-insulin-stimulated percent change activation of Akt on T308 and FOXO1/3A in the KO when compared to WT or KO mouse muscle after exercise. Our data show that a bout of treadmill exercise discriminately improves insulin-stimulated signaling in the absence of REDD1.
Collapse
Affiliation(s)
- Cory M. Dungan
- Department of Rehabilitation SciencesCollege of Health SciencesUniversity of KentuckyLexingtonKentucky
| | - Bradley S. Gordon
- Department of Nutrition, Food, and Exercise SciencesCollege of Human SciencesFlorida State UniversityTallahasseeFlorida
| | - David L. Williamson
- Kinesiology ProgramSchool of Behavioral Sciences and EducationPenn State HarrisburgMiddletownPennsylvania
| |
Collapse
|
18
|
Hypoxia impairs adaptation of skeletal muscle protein turnover- and AMPK signaling during fasting-induced muscle atrophy. PLoS One 2018; 13:e0203630. [PMID: 30212583 PMCID: PMC6136752 DOI: 10.1371/journal.pone.0203630] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/23/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Hypoxemia in humans may occur during high altitude mountaineering and in patients suffering from ventilatory insufficiencies such as cardiovascular- or respiratory disease including Chronic Obstructive Pulmonary Disease (COPD). In these conditions, hypoxemia has been correlated to reduced appetite and decreased food intake. Since hypoxemia and reduced food intake intersect in various physiological and pathological conditions and both induce loss of muscle mass, we investigated whether hypoxia aggravates fasting-induced skeletal muscle atrophy and evaluated underlying protein turnover signaling. METHODS Mice were kept under hypoxic (8% oxygen) or normoxic conditions (21% oxygen), or were pair-fed to the hypoxia group for 12 days. Following an additional 24 hours of fasting, muscle weight and protein turnover signaling were assessed in the gastrocnemius muscle by RT-qPCR and Western blotting. RESULTS Loss of gastrocnemius muscle mass in response to fasting in the hypoxic group was increased compared to the normoxic group, but not to the pair-fed normoxic control group. Conversely, the fasting-induced increase in poly-ubiquitin conjugation, and expression of the ubiquitin 26S-proteasome E3 ligases, autophagy-lysosomal degradation-related mRNA transcripts and proteins, and markers of the integrated stress response (ISR), were attenuated in the hypoxia group compared to the pair-fed group. Mammalian target of rapamycin complex 1 (mTORC1) downstream signaling was reduced by fasting under normoxic conditions, but sustained under hypoxic conditions. Activation of AMP-activated protein kinase (AMPK) / tuberous sclerosis complex 2 (TSC2) signaling by fasting was absent, in line with retained mTORC1 activity under hypoxic conditions. Similarly, hypoxia suppressed AMPK-mediated glucocorticoid receptor (GR) signaling following fasting, which corresponded with blunted proteolytic signaling responses. CONCLUSIONS Hypoxia aggravates fasting-induced muscle wasting, and suppresses AMPK and ISR activation. Altered AMPK-mediated regulation of mTORC1 and GR may underlie aberrant protein turnover signaling and affect muscle atrophy responses in hypoxic skeletal muscle.
Collapse
|
19
|
Role of mTOR in Glucose and Lipid Metabolism. Int J Mol Sci 2018; 19:ijms19072043. [PMID: 30011848 PMCID: PMC6073766 DOI: 10.3390/ijms19072043] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin, mTOR is the master regulator of a cell’s growth and metabolic state in response to nutrients, growth factors and many extracellular cues. Its dysregulation leads to a number of metabolic pathological conditions, including obesity and type 2 diabetes. Here, we review recent findings on the role of mTOR in major metabolic organs, such as adipose tissues, liver, muscle, pancreas and brain. And their potentials as the mTOR related pharmacological targets will be also discussed.
Collapse
|
20
|
Dai W, Miller WP, Toro AL, Black AJ, Dierschke SK, Feehan RP, Kimball SR, Dennis MD. Deletion of the stress-response protein REDD1 promotes ceramide-induced retinal cell death and JNK activation. FASEB J 2018; 32:fj201800413RR. [PMID: 29920218 PMCID: PMC6219834 DOI: 10.1096/fj.201800413rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The role of dyslipidemia in the development of retinal dysfunction remains poorly understood. Using an animal model of diet-induced obesity/pre-type 2 diabetes, we investigated molecular defects in the retina arising from consumption of a diet high in saturated fats and sugars ( i.e., a Western diet). We found that feeding mice a Western diet increased the abundance of retinal sphingolipids, attenuated protein kinase B (Akt) phosphorylation, enhanced JNK activation, and increased retinal cell death. When we used palmitate or C6-ceramide (Cer) to assess sphingolipid-mediated signaling in cultured murine and human cells, we observed similar effects on Akt, JNK, and cell death. Furthermore, both Western diet and C6-Cer exposure enhanced expression of the stress-response protein regulated in development and DNA damage response 1 (REDD1) and loss of REDD1 increased C6-Cer-induced JNK activation and cell death. Exogenous REDD1 expression repressed JNK-mediated phosphorylation in cultured cells. We found that thioredoxin-interacting protein (TXNIP) expression was elevated in REDD1-deficient cell lines and C6-Cer promoted TXNIP expression in both wild-type and REDD1-deficient cells. Likewise, TXNIP knockdown attenuated JNK activation and caspase 3 cleavage after either C6-Cer exposure or REDD1 deletion. The results support a model wherein Cer-induced REDD1 expression attenuates TXNIP-dependent JNK activation and retinal cell death.-Dai, W., Miller, W. P., Toro, A. L., Black, A. J., Dierschke, S. K., Feehan, R. P., Kimball, S. R., Dennis, M. D. Deletion of the stress-response protein REDD1 promotes ceramide-induced retinal cell death and JNK activation.
Collapse
Affiliation(s)
- Weiwei Dai
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - William P Miller
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Allyson L Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Adam J Black
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Sadie K Dierschke
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Robert P Feehan
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Michael D Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
21
|
Britto FA, Cortade F, Belloum Y, Blaquière M, Gallot YS, Docquier A, Pagano AF, Jublanc E, Bendridi N, Koechlin-Ramonatxo C, Chabi B, Francaux M, Casas F, Freyssenet D, Rieusset J, Giorgetti-Peraldi S, Carnac G, Ollendorff V, Favier FB. Glucocorticoid-dependent REDD1 expression reduces muscle metabolism to enable adaptation under energetic stress. BMC Biol 2018; 16:65. [PMID: 29895328 PMCID: PMC5998563 DOI: 10.1186/s12915-018-0525-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022] Open
Abstract
Background Skeletal muscle atrophy is a common feature of numerous chronic pathologies and is correlated with patient mortality. The REDD1 protein is currently recognized as a negative regulator of muscle mass through inhibition of the Akt/mTORC1 signaling pathway. REDD1 expression is notably induced following glucocorticoid secretion, which is a component of energy stress responses. Results Unexpectedly, we show here that REDD1 instead limits muscle loss during energetic stresses such as hypoxia and fasting by reducing glycogen depletion and AMPK activation. Indeed, we demonstrate that REDD1 is required to decrease O2 and ATP consumption in skeletal muscle via reduction of the extent of mitochondrial-associated endoplasmic reticulum membranes (MAMs), a central hub connecting energy production by mitochondria and anabolic processes. In fact, REDD1 inhibits ATP-demanding processes such as glycogen storage and protein synthesis through disruption of the Akt/Hexokinase II and PRAS40/mTORC1 signaling pathways in MAMs. Our results uncover a new REDD1-dependent mechanism coupling mitochondrial respiration and anabolic processes during hypoxia, fasting, and exercise. Conclusions Therefore, REDD1 is a crucial negative regulator of energy expenditure that is necessary for muscle adaptation during energetic stresses. This present study could shed new light on the role of REDD1 in several pathologies associated with energetic metabolism alteration, such as cancer, diabetes, and Parkinson’s disease. Electronic supplementary material The online version of this article (10.1186/s12915-018-0525-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | - Marine Blaquière
- PHYMEDEXP, Univ. Montpellier, INSERM, CNRS, CHRU of Montpellier, Montpellier, France
| | | | | | | | | | - Nadia Bendridi
- INSERM UMR-1060, CarMeN Laboratory, Lyon 1 University, INRA U1397, Oullins, France
| | | | | | - Marc Francaux
- Institute of Neuroscience, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | | | | | - Jennifer Rieusset
- INSERM UMR-1060, CarMeN Laboratory, Lyon 1 University, INRA U1397, Oullins, France
| | | | - Gilles Carnac
- PHYMEDEXP, Univ. Montpellier, INSERM, CNRS, CHRU of Montpellier, Montpellier, France
| | | | | |
Collapse
|
22
|
Systems Analysis of the Liver Transcriptome in Adult Male Zebrafish Exposed to the Plasticizer (2-Ethylhexyl) Phthalate (DEHP). Sci Rep 2018; 8:2118. [PMID: 29391432 PMCID: PMC5794889 DOI: 10.1038/s41598-018-20266-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/15/2018] [Indexed: 02/08/2023] Open
Abstract
The organic compound diethylhexyl phthalate (DEHP) represents a high production volume chemical found in cosmetics, personal care products, laundry detergents, and household items. DEHP, along with other phthalates causes endocrine disruption in males. Exposure to endocrine disrupting chemicals has been linked to the development of several adverse health outcomes with apical end points including Non-Alcoholic Fatty Liver Disease (NAFLD). This study examined the adult male zebrafish (Danio rerio) transcriptome after exposure to environmental levels of DEHP and 17α-ethinylestradiol (EE2) using both DNA microarray and RNA-sequencing technologies. Our results show that exposure to DEHP is associated with differentially expressed (DE) transcripts associated with the disruption of metabolic processes in the liver, including perturbation of five biological pathways: ‘FOXA2 and FOXA3 transcription factor networks’, ‘Metabolic pathways’, ‘metabolism of amino acids and derivatives’, ‘metabolism of lipids and lipoproteins’, and ‘fatty acid, triacylglycerol, and ketone body metabolism’. DE transcripts unique to DEHP exposure, not observed with EE2 (i.e. non-estrogenic effects) exhibited a signature related to the regulation of transcription and translation, and ruffle assembly and organization. Collectively our results indicate that exposure to low DEHP levels modulates the expression of liver genes related to fatty acid metabolism and the development of NAFLD.
Collapse
|
23
|
Gordon BS, Steiner JL, Rossetti ML, Qiao S, Ellisen LW, Govindarajan SS, Eroshkin AM, Williamson DL, Coen PM. REDD1 induction regulates the skeletal muscle gene expression signature following acute aerobic exercise. Am J Physiol Endocrinol Metab 2017; 313:E737-E747. [PMID: 28899858 PMCID: PMC5814598 DOI: 10.1152/ajpendo.00120.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/08/2017] [Accepted: 09/04/2017] [Indexed: 11/22/2022]
Abstract
The metabolic stress placed on skeletal muscle by aerobic exercise promotes acute and long-term health benefits in part through changes in gene expression. However, the transducers that mediate altered gene expression signatures have not been completely elucidated. Regulated in development and DNA damage 1 (REDD1) is a stress-induced protein whose expression is transiently increased in skeletal muscle following acute aerobic exercise. However, the role of this induction remains unclear. Because REDD1 altered gene expression in other model systems, we sought to determine whether REDD1 induction following acute exercise altered the gene expression signature in muscle. To do this, wild-type and REDD1-null mice were randomized to remain sedentary or undergo a bout of acute treadmill exercise. Exercised mice recovered for 1, 3, or 6 h before euthanization. Acute exercise induced a transient increase in REDD1 protein expression within the plantaris only at 1 h postexercise, and the induction occurred in both cytosolic and nuclear fractions. At this time point, global changes in gene expression were surveyed using microarray. REDD1 induction was required for the exercise-induced change in expression of 24 genes. Validation by RT-PCR confirmed that the exercise-mediated changes in genes related to exercise capacity, muscle protein metabolism, neuromuscular junction remodeling, and Metformin action were negated in REDD1-null mice. Finally, the exercise-mediated induction of REDD1 was partially dependent upon glucocorticoid receptor activation. In all, these data show that REDD1 induction regulates the exercise-mediated change in a distinct set of genes within skeletal muscle.
Collapse
Affiliation(s)
- Bradley S Gordon
- Department of Nutrition, Food, and Exercise Science, Florida State University, Tallahassee, Florida;
- Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, Florida
| | - Jennifer L Steiner
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Michael L Rossetti
- Department of Nutrition, Food, and Exercise Science, Florida State University, Tallahassee, Florida
- Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, Florida
| | - Shuxi Qiao
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Leif W Ellisen
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | - Alexey M Eroshkin
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - David L Williamson
- Kinesiology Program, School of Behavioral Sciences and Education, Pennsylvania State University-Harrisburg, Middletown, Pennsylvania; and
| | - Paul M Coen
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| |
Collapse
|
24
|
Faecalibacterium prausnitzii treatment improves hepatic health and reduces adipose tissue inflammation in high-fat fed mice. ISME JOURNAL 2017; 11:1667-1679. [PMID: 28375212 DOI: 10.1038/ismej.2017.24] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/10/2016] [Accepted: 01/27/2017] [Indexed: 02/06/2023]
Abstract
Faecalibacterium prausnitzii is considered as one of the most important bacterial indicators of a healthy gut. We studied the effects of oral F. prausnitzii treatment on high-fat fed mice. Compared to the high-fat control mice, F. prausnitzii-treated mice had lower hepatic fat content, aspartate aminotransferase and alanine aminotransferase, and increased fatty acid oxidation and adiponectin signaling in liver. Hepatic lipidomic analyses revealed decreases in several species of triacylglycerols, phospholipids and cholesteryl esters. Adiponectin expression was increased in the visceral adipose tissue, and the subcutaneous and visceral adipose tissues were more insulin sensitive and less inflamed in F. prausnitzii-treated mice. Further, F. prausnitzii treatment increased muscle mass that may be linked to enhanced mitochondrial respiration, modified gut microbiota composition and improved intestinal integrity. Our findings show that F. prausnitzii treatment improves hepatic health, and decreases adipose tissue inflammation in mice and warrant the need for further studies to discover its therapeutic potential.
Collapse
|
25
|
Lipina C, Hundal HS. Is REDD1 a Metabolic Éminence Grise? Trends Endocrinol Metab 2016; 27:868-880. [PMID: 27613400 PMCID: PMC5119498 DOI: 10.1016/j.tem.2016.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/04/2016] [Accepted: 08/09/2016] [Indexed: 01/31/2023]
Abstract
Regulated in development and DNA damage response 1 (REDD1) has been functionally linked to the control of diverse cellular processes due, at least in part, to its ability to repress mammalian or mechanistic Target of Rapamycin (mTOR) Complex-1 (mTORC1), a key protein complex controlled by hormonal and nutrient cues. Notably, emerging evidence suggests that REDD1 also regulates several pathways involved in modulating energy balance and metabolism. Herein, we discuss evidence implicating REDD1 as a key modulator of insulin action and metabolic function, including its potential contribution to mitochondrial biology and pancreatic islet function. Collectively, the available evidence suggests that REDD1 has a more prominent role in energy homeostasis than was previously thought, and implicates REDD1 as a potential therapeutic target for treatment of metabolic disorders.
Collapse
Affiliation(s)
- Christopher Lipina
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Harinder S Hundal
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| |
Collapse
|
26
|
Regulation of skeletal muscle insulin-stimulated signaling through the MEK-REDD1-mTOR axis. Biochem Biophys Res Commun 2016; 482:1067-1072. [PMID: 27913296 DOI: 10.1016/j.bbrc.2016.11.159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 11/28/2016] [Indexed: 01/22/2023]
Abstract
Recent findings in adipocytes suggest that mitogen-activated protein kinase (MAPK)/extracellular-regulated signaling kinase (ERK) kinase 1/2 (MEK1/2) signaling regulates regulated in development and DNA damage 1 (REDD1) protein expression. Similarly, our previous work show that a lack of REDD1 protein expression, and associated hyperactive basal mechanistic target of rapamycin (mTOR) signaling, limits skeletal muscle's response to insulin. Therefore, we sought to determine: 1) if MEK1/2 inhibition is sufficient to reduce REDD1 protein expression and subsequently insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation via negative feedback of hyperactive mTOR in REDD1 wild-type (WT) mice and 2) if rapamycin-mediated mTOR inhibition is sufficient to improve IRS-1 tyrosine phosphorylation in REDD1 knockout (KO) mice. REDD1 WT mice were injected with 10 mg/kg BW of the MEK1/2 non-competitive inhibitor, PD184352, 3 h prior to acute insulin treatment. In separate studies, REDD1 KO mice were injected with 5 mg/kg BW of the mTOR inhibitor, rapamycin, 3 h prior to acute insulin treatment. Following the inhibitor treatment period, markers of insulin signaling activation (IRS-1 Y1222, MEK1/2 S217/221, ERK1/2 T202/Y204), REDD1, and mTOR signaling activation (S6K1 T389, rpS6 S240/244) were examined in skeletal muscle collected before and after a 10 min insulin treatment. PD184352 treatment reduced MEK/ERK phosphorylation and REDD1 protein expression, independent of insulin. This reduction in REDD1 protein expression was associated with elevated basal S6K1 and rpS6 phosphorylation and reduced insulin stimulated IRS-1 phosphorylation. Conversely, rapamycin inhibited S6K1 and rpS6 activation, and significantly improved insulin -stimulated activation of IRS-1 and MEK1/2 in KO mice. These data support that REDD1 is required for normal insulin-stimulated signaling, and that a subtle balance exists between MEK1/2, REDD1, and mTOR for the proper regulation of insulin signaling.
Collapse
|
27
|
Beals JW, Sukiennik RA, Nallabelli J, Emmons RS, van Vliet S, Young JR, Ulanov AV, Li Z, Paluska SA, De Lisio M, Burd NA. Anabolic sensitivity of postprandial muscle protein synthesis to the ingestion of a protein-dense food is reduced in overweight and obese young adults. Am J Clin Nutr 2016; 104:1014-1022. [PMID: 27604771 DOI: 10.3945/ajcn.116.130385] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 08/01/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Excess body fat diminishes muscle protein synthesis rates in response to hyperinsulinemic-hyperaminoacidemic clamps. However, muscle protein synthetic responses after the ingestion of a protein-dense food source across a range of body mass indexes (BMIs) have not been compared. OBJECTIVE We compared the myofibrillar protein synthetic response and underlying nutrient-sensing mechanisms after the ingestion of lean pork between obese, overweight, and healthy-weight adults. DESIGN Ten healthy-weight [HW; BMI (in kg/m2): 22.7 ± 0.4], 10 overweight (OW; BMI: 27.1 ± 0.5), and 10 obese (OB; BMI: 35.9 ± 1.3) adults received primed continuous l-[ring-13C6]phenylalanine infusions. Blood and muscle biopsy samples were collected before and after the ingestion of 170 g pork (36 g protein and 3 g fat) to assess skeletal muscle anabolic signaling, amino acid transporters [large neutral and small neutral amino acid transporters (LAT1, SNAT2) and CD98], and myofibrillar protein synthesis. RESULTS At baseline, OW and OB groups showed greater relative amounts of mammalian target of rapamycin complex 1 (mTORC1) protein than the HW group. Pork ingestion increased mTORC1 phosphorylation only in the HW group (P = 0.001). LAT1 and SNAT2 protein content increased during the postprandial period in all groups (time effect, P < 0.05). Basal myofibrillar protein synthetic responses were similar between groups (P = 0.43). However, myofibrillar protein synthetic responses (0-300 min) were greater in the HW group (1.6-fold; P = 0.005) after pork ingestion than in the OW and OB groups. CONCLUSIONS There is a diminished myofibrillar protein synthetic response to the ingestion of protein-dense food in overweight and obese adults compared with healthy-weight controls. These data indicate that impaired postprandial myofibrillar protein synthetic response may be an early defect with increasing fat mass, potentially dependent on altered anabolic signals, that reduces muscle sensitivity to food ingestion. This trial was registered at clinicaltrials.gov as NCT02613767.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhong Li
- Roy J Carver Biotechnology Center, and
| | - Scott A Paluska
- Department of Family Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Michael De Lisio
- Division of Nutritional Sciences, Department of Kinesiology and Community Health
| | - Nicholas A Burd
- Division of Nutritional Sciences, Department of Kinesiology and Community Health,
| |
Collapse
|
28
|
Roy B, Curtis ME, Fears LS, Nahashon SN, Fentress HM. Molecular Mechanisms of Obesity-Induced Osteoporosis and Muscle Atrophy. Front Physiol 2016; 7:439. [PMID: 27746742 PMCID: PMC5040721 DOI: 10.3389/fphys.2016.00439] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 09/15/2016] [Indexed: 12/19/2022] Open
Abstract
Obesity and osteoporosis are two alarming health disorders prominent among middle and old age populations, and the numbers of those affected by these two disorders are increasing. It is estimated that more than 600 million adults are obese and over 200 million people have osteoporosis worldwide. Interestingly, both of these abnormalities share some common features including a genetic predisposition, and a common origin: bone marrow mesenchymal stromal cells. Obesity is characterized by the expression of leptin, adiponectin, interleukin 6 (IL-6), interleukin 10 (IL-10), monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), macrophage colony stimulating factor (M-CSF), growth hormone (GH), parathyroid hormone (PTH), angiotensin II (Ang II), 5-hydroxy-tryptamine (5-HT), Advance glycation end products (AGE), and myostatin, which exert their effects by modulating the signaling pathways within bone and muscle. Chemical messengers (e.g., TNF-α, IL-6, AGE, leptins) that are upregulated or downregulated as a result of obesity have been shown to act as negative regulators of osteoblasts, osteocytes and muscles, as well as positive regulators of osteoclasts. These additive effects of obesity ultimately increase the risk for osteoporosis and muscle atrophy. The aim of this review is to identify the potential cellular mechanisms through which obesity may facilitate osteoporosis, muscle atrophy and bone fractures.
Collapse
Affiliation(s)
- Bipradas Roy
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Mary E Curtis
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Letimicia S Fears
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Samuel N Nahashon
- Department of Agricultural and Environmental Sciences, Tennessee State University Nashville, TN, USA
| | - Hugh M Fentress
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| |
Collapse
|
29
|
Gordon BS, Steiner JL, Williamson DL, Lang CH, Kimball SR. Emerging role for regulated in development and DNA damage 1 (REDD1) in the regulation of skeletal muscle metabolism. Am J Physiol Endocrinol Metab 2016; 311:E157-74. [PMID: 27189933 PMCID: PMC4967146 DOI: 10.1152/ajpendo.00059.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/11/2016] [Indexed: 12/25/2022]
Abstract
Since its discovery, the protein regulated in development and DNA damage 1 (REDD1) has been implicated in the cellular response to various stressors. Most notably, its role as a repressor of signaling through the central metabolic regulator, the mechanistic target of rapamycin in complex 1 (mTORC1) has gained considerable attention. Not surprisingly, changes in REDD1 mRNA and protein have been observed in skeletal muscle under various physiological conditions (e.g., nutrient consumption and resistance exercise) and pathological conditions (e.g., sepsis, alcoholism, diabetes, obesity) suggesting a role for REDD1 in regulating mTORC1-dependent skeletal muscle protein metabolism. Our understanding of the causative role of REDD1 in skeletal muscle metabolism is increasing mostly due to the availability of genetically modified mice in which the REDD1 gene is disrupted. Results from such studies provide support for an important role for REDD1 in the regulation of mTORC1 as well as reveal unexplored functions of this protein in relation to other aspects of skeletal muscle metabolism. The goal of this work is to provide a comprehensive review of the role of REDD1 (and its paralog REDD2) in skeletal muscle during both physiological and pathological conditions.
Collapse
Affiliation(s)
- Bradley S Gordon
- Institute of Exercise Physiology and Wellness, The University of Central Florida, Orlando, Florida;
| | - Jennifer L Steiner
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and
| | - David L Williamson
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and
| |
Collapse
|
30
|
Caloric Restriction Normalizes Obesity-Induced Alterations on Regulators of Skeletal Muscle Growth Signaling. Lipids 2016; 51:905-12. [PMID: 27289530 DOI: 10.1007/s11745-016-4168-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/17/2016] [Indexed: 10/21/2022]
Abstract
The objective of this study was to establish the impact of caloric restriction on high fat diet-induced alterations on regulators of skeletal muscle growth. We hypothesized that caloric restriction would reverse the negative effects of high fat diet-induced obesity on REDD1 and mTOR-related signaling. Following an initial 8 week period of HF diet-induced obesity, caloric restriction (CR ~30 %) was employed while mice continued to consume either a low (LF) or high fat (HF) diet for 8 weeks. Western analysis of skeletal muscle showed that CR reduced (p < 0.05) the obesity-related effects on the lipogenic protein, SREBP1. Likewise, CR reduced (p < 0.05) the obesity-related effects on the hyperactivation of mTORC1 and ERK1/2 signaling to levels comparable to the LF mice. CR also reduced (p < 0.05) obesity-induced expression of negative regulators of growth, REDD1 and cleaved caspase 3. These findings have implications for on the reversibility of dysregulated growth signaling in obese skeletal muscle, using short-term caloric restriction.
Collapse
|
31
|
Ray AD, Personius KE, Williamson DL, Dungan CM, Dhillon SS, Hershberger PA. Vitamin D3 intake modulates diaphragm but not peripheral muscle force in young mice. J Appl Physiol (1985) 2016; 120:1124-31. [PMID: 26968027 DOI: 10.1152/japplphysiol.00643.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 03/09/2016] [Indexed: 12/16/2022] Open
Abstract
Recent data support an important role for vitamin D in respiratory health. We tested the hypothesis that dietary vitamin D3 (VD3) intake modulates diaphragm (DIA) strength. Four-week-old female A/J mice (n = 10/group) were randomized to receive diets containing 100 IU VD3/kg (low), 1,000 IU VD3/kg (reference), or 10,000 IU VD3/kg (pharmacologic). After 6 wk of dietary intervention, plasma 25-hydroxyvitamin D3 (25D3) levels, DIA and extensor digitorum longus (EDL) in vitro contractile properties, and fiber cross-sectional area (CSA) were measured. Myosin heavy chain (MHC) composition and Akt/Foxo3A growth signaling were studied in the DIA and tibialis anterior. Mice fed the low, reference, and pharmacologic diets had average 25D3 levels of 7, 21, and 59 ng/ml, respectively. Maximal DIA force, twitch force, and fiber CSA were reduced 26%, 28%, and 10% (P < 0.01), respectively, in mice receiving the low-VD3 diet compared with the reference and pharmacologic diets. EDL force parameters were unaltered by diet. Effects of VD3 intake on DIA force were not observed in mice that began dietary intervention at 12 wk of age. VD3 intake did not alter the MHC composition of the DIA, indicating that decreases in force and CSA in young mice were not due to a switch in fiber type. Paradoxically, low VD3 intake was associated with activation of anabolic signaling in muscle (hyperphosphorylation of Akt and Foxo3A and decreased expression of autophagy marker LC3). These studies identify a potential role of dietary VD3 in regulating DIA development and insulin sensitivity.
Collapse
Affiliation(s)
- Andrew D Ray
- Department of Rehabilitation Science, University at Buffalo, Buffalo, New York;
| | | | - David L Williamson
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York
| | - Cory M Dungan
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York
| | - Samjot S Dhillon
- Department of Medicine, Thoracic Oncology, Roswell Park Cancer Institute, Buffalo, New York; and
| | - Pamela A Hershberger
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
32
|
Steiner JL, Crowell KT, Kimball SR, Lang CH. Disruption of REDD1 gene ameliorates sepsis-induced decrease in mTORC1 signaling but has divergent effects on proteolytic signaling in skeletal muscle. Am J Physiol Endocrinol Metab 2015; 309:E981-94. [PMID: 26487002 PMCID: PMC4816198 DOI: 10.1152/ajpendo.00264.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/14/2015] [Indexed: 01/04/2023]
Abstract
Sepsis-induced skeletal muscle atrophy and weakness are due in part to decreased mTORC1-mediated protein synthesis and increased proteolysis via the autophagy-lysosomal system and ubiquitin-proteasome pathway. The REDD1 (regulated in development and DNA damage-1) protein is increased in sepsis and can negatively regulate mTORC1 activity. However, the contribution of REDD1 to the sepsis-induced change in muscle protein synthesis and degradation has not been determined. Sepsis was produced by cecal ligation and puncture in female REDD1(-/-) or wild-type (WT) mice, and end points were assessed 24 h later in gastrocnemius; time-matched, pair-fed controls of each genotype were included. Sepsis increased REDD1 protein 300% in WT mice, whereas REDD1 was absent in REDD1(-/-) muscle. Sepsis decreased protein synthesis and phosphorylation of downstream targets of mTORC1 (S6K1 Thr(389), rpS6 Ser(240/244), 4E-BP1 Ser(65)) in WT but not REDD1(-/-) mice. However, Akt and PRAS40 phosphorylation was suppressed in both sham and septic muscle from REDD1(-/-) mice despite unaltered PDK1, PP2A, or TSC2 expression. Sepsis increased autophagy as indicated by decreased ULK1 Ser(757) phosphorylation and p62 abundance and increased LC3B-II/I in WT mice, whereas these changes were absent in septic REDD1(-/-) mice. Conversely, REDD1 deletion did not prevent the sepsis-induced decrease in IGF-I mRNA or the concomitant increase in IL-6, TNFα, MuRF1, and atrogin1 mRNA expression. Lastly, 5-day survival in a separate set of septic mice did not differ between WT and REDD1(-/-) mice. These data highlight the central role of REDD1 in regulating both protein synthesis and autophagy in skeletal muscle during sepsis.
Collapse
Affiliation(s)
| | - Kristen T Crowell
- Department of Cellular and Molecular Physiology and Department of Surgery, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | | | - Charles H Lang
- Department of Cellular and Molecular Physiology and Department of Surgery, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
33
|
Williamson DL, Dungan CM, Mahmoud AM, Mey JT, Blackburn BK, Haus JM. Aberrant REDD1-mTORC1 responses to insulin in skeletal muscle from Type 2 diabetics. Am J Physiol Regul Integr Comp Physiol 2015; 309:R855-63. [PMID: 26269521 PMCID: PMC4666944 DOI: 10.1152/ajpregu.00285.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/04/2015] [Indexed: 11/22/2022]
Abstract
The objective of this study was to establish whether alterations in the REDD1-mTOR axis underlie skeletal muscle insensitivity to insulin in Type 2 diabetic (T2D), obese individuals. Vastus lateralis muscle biopsies were obtained from lean, control and obese, T2D subjects under basal and after a 2-h hyperinsulinemic (40 mU·m(-2)·min(-1))-euglycemic (5 mM) clamp. Muscle lysates were examined for total REDD1, and phosphorylated Akt, S6 kinase 1 (S6K1), 4E-BP1, ERK1/2, and MEK1/2 via Western blot analysis. Under basal conditions [(-) insulin], T2D muscle exhibited higher S6K1 and ERK1/2 and lower 4E-BP1 phosphorylation (P < 0.05), as well as elevations in blood cortisol, glucose, insulin, glycosylated hemoglobin (P < 0.05) vs. lean controls. Following insulin infusion, whole body glucose disposal rates (GDR; mg/kg/min) were lower (P < 0.05) in the T2D vs. the control group. The basal-to-insulin percent change in REDD1 expression was higher (P < 0.05) in muscle from the T2D vs. the control group. Whereas, the basal-to-insulin percent change in muscle Akt, S6K1, ERK1/2, and MEK1/2 phosphorylation was significantly lower (P < 0.05) in the T2D vs. the control group. Findings from this study propose a REDD1-regulated mechanism in T2D skeletal muscle that may contribute to whole body insulin resistance and may be a target to improve insulin action in insulin-resistant individuals.
Collapse
Affiliation(s)
- David L Williamson
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York; and
| | - Cory M Dungan
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York; and
| | - Abeer M Mahmoud
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Jacob T Mey
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Brian K Blackburn
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Jacob M Haus
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
34
|
Steiner JL, Kimball SR, Lang CH. Acute Alcohol-Induced Decrease in Muscle Protein Synthesis in Female Mice Is REDD-1 and mTOR-Independent. Alcohol Alcohol 2015; 51:242-50. [PMID: 26394774 DOI: 10.1093/alcalc/agv105] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/26/2015] [Indexed: 12/20/2022] Open
Abstract
AIMS To determine the causative role of the REDD (regulated in development and DNA damage)-1 protein, a known negative regulator of mTOR kinase, in changes in muscle protein synthesis induced by acute alcohol administration. METHODS Adult female REDD1(-/-) or wild-type (WT) mice were injected IP with ethanol (alcohol; 3 g/kg BW) or saline and the skeletal muscle was removed 1 h later. In vivo protein synthesis was assessed as were selected endpoints related to the activation of mTOR and protein degradation. RESULTS Acute alcohol decreased muscle protein synthesis similarly in WT and REDD1(-/-) mice. In contrast, mTORC1 signaling was largely unaffected by either EtOH or genotype as evidenced by the lack of change in the phosphorylation of its downstream targets, S6K1 T(389) and 4E-BP1 S(65). Although alcohol decreased p62 and ULK1 S(757) protein in muscle from WT and REDD1(-/-) mice, there was no change in LC3B lipidation, or beclin1, Atg7 and Atg12 protein suggesting no change in autophagy. MuRF1 and atrogin-1 mRNAs were elevated in alcohol-treated REDD1(-/-) mice compared with WT mice suggesting activation of the ubiquitin proteasome activity. While there was no genotype or alcohol effect on plasma corticosterone, REDD1(-/-) mice failed to demonstrate the alcohol-induced hyperinsulinemia seen in WT mice. CONCLUSION REDD1 does not appear to play a role in the acute alcohol-mediated decrease in protein synthesis or mTOR activity, but may contribute to the regulation of ubiquitin-proteasome mediated protein breakdown.
Collapse
Affiliation(s)
- Jennifer L Steiner
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| |
Collapse
|
35
|
The bigger, the stronger? Insights from muscle architecture and nervous characteristics in obese adolescent girls. Int J Obes (Lond) 2015; 40:245-51. [PMID: 26285605 DOI: 10.1038/ijo.2015.158] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 07/08/2015] [Accepted: 08/03/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND Young obese youth are generally stronger than lean youth. This has been linked to the loading effect of excess body mass, acting as a training stimulus comparable to strength training. Whether this triggers specific adaptations of the muscle architecture (MA) and voluntary activation (VA) that could account for the higher strength of obese subjects remains unknown. METHODS MA characteristics (that is, pennation angle (PA), fascicle length (FL) and muscle thickness (MT)) and muscle size (that is, anatomical cross-sectional area (ACSA)) of the knee extensor (KE) and plantar flexor (PF) muscles were evaluated in 12 obese and 12 non-obese adolescent girls (12-15 years). Maximal isometric torque and VA of the KE and PF muscles were also assessed. RESULTS Results revealed higher PA (P<0.05), greater MT (P<0.001), ACSA (P<0.01), segmental lean mass (P<0.001) and VA (P<0.001) for KE and PF muscles in obese girls. Moreover, obese individuals produced a higher absolute torque than their lean counterparts on the KE (224.6±39.5 vs 135.7±32.7 N m, respectively; P<0.001) and PF muscles (73.3±16.5 vs 44.5±6.2 N m; P<0.001). Maximal voluntary contraction (MVC) was correlated to PA for the KE (r=0.46-0.57, P<0.05-0.01) and PF muscles (r=0.45-0.55, P<0.05-0.01). MVC was also correlated with VA (KE: r=0.44, P<0.05; PF: r=0.65, P<0.001) and segmental lean mass (KE: r=0.48, P<0.05; PF: r=0.57, P<0.01). CONCLUSIONS This study highlighted favorable muscular and nervous adaptations to obesity that account for the higher strength of obese youth. The excess of body mass supported during daily activities could act as a chronic training stimulus responsible for these adaptations.
Collapse
|
36
|
Gordon BS, Williamson DL, Lang CH, Jefferson LS, Kimball SR. Nutrient-induced stimulation of protein synthesis in mouse skeletal muscle is limited by the mTORC1 repressor REDD1. J Nutr 2015; 145:708-13. [PMID: 25716553 PMCID: PMC4381770 DOI: 10.3945/jn.114.207621] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 01/29/2015] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND In skeletal muscle, the nutrient-induced stimulation of protein synthesis requires signaling through the mechanistic target of rapamycin complex 1 (mTORC1). Expression of the repressor of mTORC1 signaling, regulated in development and DNA damage 1 (REDD1), is elevated in muscle during various atrophic conditions and diminished under hypertrophic conditions. The question arises as to what extent REDD1 limits the nutrient-induced stimulation of protein synthesis. OBJECTIVE The objective was to examine the role of REDD1 in limiting the response of muscle protein synthesis and mTORC1 signaling to a nutrient stimulus. METHODS Wild type REDD1 gene (REDD1(+/+)) and disruption in the REDD1 gene (REDD1(-/-)) mice were feed deprived for 16 h and randomized to remain feed deprived or refed for 15 or 60 min. The tibialis anterior was then removed for analysis of protein synthesis and mTORC1 signaling. RESULTS In feed-deprived mice, protein synthesis and mTORC1 signaling were significantly lower in REDD1(+/+) than in REDD1(-/-) mice. Thirty minutes after the start of refeeding, protein synthesis in REDD1(+/+) mice was stimulated by 28%, reaching a value similar to that observed in feed-deprived REDD1(-/-) mice, and was accompanied by increased phosphorylation of mTOR (Ser2448), p70S6K1 (Thr389), and 4E-BP1 (Ser65) by 81%, 167%, and 207%, respectively. In refed REDD1(-/-) mice, phosphorylation of mTOR (Ser2448), p70S6K1 (Thr389), and 4E-BP1 (Ser65) were significantly augmented above the values observed in refed REDD1(+/+) mice by 258%, 405%, and 401%, respectively, although protein synthesis was not coordinately increased. Seventy-five minutes after refeeding, REDD1 expression in REDD1(+/+) mice was reduced (∼15% of feed-deprived REDD1(+/+) values), and protein synthesis and mTORC1 signaling were not different between refed REDD1(+/+) mice and REDD1(-/-) mice. CONCLUSIONS The results show that REDD1 expression limits protein synthesis in mouse skeletal muscle by inhibiting mTORC1 signaling during periods of feed deprivation and that a reduction in its expression is necessary for maximal stimulation of protein synthesis in response to refeeding.
Collapse
Affiliation(s)
- Bradley S Gordon
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA; and
| | - David L Williamson
- Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, NY
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA; and
| | - Leonard S Jefferson
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA; and
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA; and
| |
Collapse
|
37
|
Britto FA, Begue G, Rossano B, Docquier A, Vernus B, Sar C, Ferry A, Bonnieu A, Ollendorff V, Favier FB. REDD1 deletion prevents dexamethasone-induced skeletal muscle atrophy. Am J Physiol Endocrinol Metab 2014; 307:E983-93. [PMID: 25315696 DOI: 10.1152/ajpendo.00234.2014] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
REDD1 (regulated in development and DNA damage response 1) has been proposed to inhibit the mechanistic target of rapamycin complex 1 (mTORC1) during in vitro hypoxia. REDD1 expression is low under basal conditions but is highly increased in response to several catabolic stresses, like hypoxia and glucocorticoids. However, REDD1 function seems to be tissue and stress dependent, and its role in skeletal muscle in vivo has been poorly characterized. Here, we investigated the effect of REDD1 deletion on skeletal muscle mass, protein synthesis, proteolysis, and mTORC1 signaling pathway under basal conditions and after glucocorticoid administration. Whereas skeletal muscle mass and typology were unchanged between wild-type (WT) and REDD1-null mice, oral gavage with dexamethasone (DEX) for 7 days reduced tibialis anterior and gastrocnemius muscle weights as well as tibialis anterior fiber size only in WT. Similarly, REDD1 deletion prevented the inhibition of protein synthesis and mTORC1 activity (assessed by S6, 4E-BP1, and ULK1 phosphorylation) observed in gastrocnemius muscle of WT mice following single DEX administration for 5 h. However, our results suggest that REDD1-mediated inhibition of mTORC1 in skeletal muscle is not related to the modulation of the binding between TSC2 and 14-3-3. In contrast, our data highlight a new mechanism involved in mTORC1 inhibition linking REDD1, Akt, and PRAS40. Altogether, these results demonstrated in vivo that REDD1 is required for glucocorticoid-induced inhibition of protein synthesis via mTORC1 downregulation. Inhibition of REDD1 may thus be a strategy to limit muscle loss in glucocorticoid-mediated atrophy.
Collapse
Affiliation(s)
- Florian A Britto
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Gwenaelle Begue
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Bernadette Rossano
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Aurélie Docquier
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Barbara Vernus
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Chamroeun Sar
- Institut National de la Sante et de la Recherche Medicale (INSERM) U 583, Institut de Neuroscience de Montpellier, France
| | - Arnaud Ferry
- Institut de Myologie, INSERM, U974, Centre National de la Recherche Scientifique UMR 7215, Université Pierre et Marie Curie, Paris, France; and Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Bonnieu
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - Vincent Ollendorff
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France
| | - François B Favier
- Institut National de la Recherche Agronomique, UMR 866 Dynamique Musculaire et Métabolisme, F-34000 Montpellier, France; Université Montpellier 1, F-34000 Montpellier, France; Université Montpellier 2, F-34000 Montpellier, France;
| |
Collapse
|
38
|
Li Z, Dungan CM, Carrier B, Rideout TC, Williamson DL. Alpha-lipoic acid supplementation reduces mTORC1 signaling in skeletal muscle from high fat fed, obese Zucker rats. Lipids 2014; 49:1193-201. [PMID: 25366515 DOI: 10.1007/s11745-014-3964-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/13/2014] [Indexed: 12/27/2022]
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway is hyperactive in liver, adipose and skeletal muscle tissues of obese rodents. Alpha-lipoic acid (αLA) has been well accepted as a weight-loss treatment, though there are limited studies on its effect on mTOR signaling in high-fat fed, obese rodents. Therefore, the goal of this study was to determine mTOR signaling and oxidative protein alterations in skeletal muscle of high-fat fed, obese rats after αLA supplementation. Phosphorylation of the mTOR substrate, eukaryotic initiation factor (eIF) 4E-binding protein 1 (4E-BP1) and eIF4B were significantly reduced (p < 0.05) in muscle from αLA supplemented rats. Activation of AMP-activated protein kinase (AMPK), an mTOR inhibitory kinase, was higher (p < 0.05) in the αLA group. Protein expression of markers of oxidative metabolism, acetyl CoA carboxylase (ACC), cytochrome c oxidase IV (COX IV), peroxisome proliferator-activated receptor (PPAR), and PPAR gamma coactivator 1-alpha (PGC-1α) were significantly higher (p < 0.05) after αLA supplementation compared to non-supplemented group. Our findings show that αLA supplementation limits the negative ramifications of consuming a high fat diet on skeletal muscle markers of oxidative metabolism and mTORC1 signaling.
Collapse
Affiliation(s)
- Zhuyun Li
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, SUNY, 2 Sherman Hall (Office)/5 Sherman Hall (Lab), Buffalo, NY, 14214, USA
| | | | | | | | | |
Collapse
|
39
|
Gordon BS, Steiner JL, Lang CH, Jefferson LS, Kimball SR. Reduced REDD1 expression contributes to activation of mTORC1 following electrically induced muscle contraction. Am J Physiol Endocrinol Metab 2014; 307:E703-11. [PMID: 25159324 PMCID: PMC4200302 DOI: 10.1152/ajpendo.00250.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Regulated in DNA damage and development 1 (REDD1) is a repressor of mTOR complex 1 (mTORC1) signaling. In humans, REDD1 mRNA expression in skeletal muscle is repressed following resistance exercise in association with activation of mTORC1. However, whether REDD1 protein expression is also reduced after exercise and if so to what extent the loss contributes to exercise-induced activation of mTORC1 is unknown. Thus, the purpose of the present study was to examine the role of REDD1 in governing the response of mTORC1 and protein synthesis to a single bout of muscle contractions. Eccentric contractions of the tibialis anterior were elicited via electrical stimulation of the sciatic nerve in male mice in either the fasted or fed state or in fasted wild-type or REDD1-null mice. Four hours postcontractions, mTORC1 signaling and protein synthesis were elevated in fasted mice in association with repressed REDD1 expression relative to nonstimulated controls. Feeding coupled with contractions further elevated mTORC1 signaling, whereas REDD1 protein expression was repressed compared with either feeding or contractions alone. Basal mTORC1 signaling and protein synthesis were elevated in REDD1-null compared with wild-type mice. The magnitude of the increase in mTORC1 signaling was similar in both wild-type and REDD1-null mice, but, unlike wild-type mice, muscle contractions did not stimulate protein synthesis in mice deficient for REDD1, presumably because basal rates were already elevated. Overall, the data demonstrate that REDD1 expression contributes to the modulation of mTORC1 signaling following feeding- and contraction-induced activation of the pathway.
Collapse
Affiliation(s)
- Bradley S Gordon
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jennifer L Steiner
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Leonard S Jefferson
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
40
|
Dungan CM, Wright DC, Williamson DL. Lack of REDD1 reduces whole body glucose and insulin tolerance, and impairs skeletal muscle insulin signaling. Biochem Biophys Res Commun 2014; 453:778-83. [PMID: 25445588 DOI: 10.1016/j.bbrc.2014.10.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/07/2014] [Indexed: 12/21/2022]
Abstract
A lack of the REDD1 promotes dysregulated growth signaling, though little has been established with respect to the metabolic role of REDD1. Therefore, the goal of this study was to determine the role of REDD1 on glucose and insulin tolerance, as well as insulin stimulated growth signaling pathway activation in skeletal muscle. First, intraperitoneal (IP) injection of glucose or insulin were administered to REDD1 wildtype (WT) versus knockout (KO) mice to examine changes in blood glucose over time. Next, alterations in skeletal muscle insulin (IRS-1, Akt, ERK 1/2) and growth (4E-BP1, S6K1, REDD1) signaling intermediates were determined before and after IP insulin treatment (10min). REDD1 KO mice were both glucose and insulin intolerant when compared to WT mice, evident by higher circulating blood glucose concentrations and a greater area under the curve following IP injections of glucose or insulin. While the REDD1 KO exhibited significant though blunted insulin-stimulated increases (p<0.05) in Akt S473 and T308 phosphorylation versus the WT mice, acute insulin treatment has no effect (p<0.05) on REDD1 KO skeletal muscle 4E-BP1 T37/46, S6K1 T389, IRS-1 Y1222, and ERK 1/2 T202/Y204 phosphorylation versus the WT mice. Collectively, these novel data suggest that REDD1 has a more distinct role in whole body and skeletal muscle metabolism and insulin action than previously thought.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States
| | - David C Wright
- Department of Human Health and Nutrition Sciences, University of Guelph, Guelph, ON, Canada
| | - David L Williamson
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, United States.
| |
Collapse
|