101
|
Chen L, Liao B, Qi H, Xie LJ, Huang L, Tan WJ, Zhai N, Yuan LB, Zhou Y, Yu LJ, Chen QF, Shu W, Xiao S. Autophagy contributes to regulation of the hypoxia response during submergence in Arabidopsis thaliana. Autophagy 2016; 11:2233-46. [PMID: 26566261 PMCID: PMC4835207 DOI: 10.1080/15548627.2015.1112483] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Autophagy involves massive degradation of intracellular components and functions as a conserved system that helps cells to adapt to adverse conditions. In mammals, hypoxia rapidly stimulates autophagy as a cell survival response. Here, we examine the function of autophagy in the regulation of the plant response to submergence, an abiotic stress that leads to hypoxia and anaerobic respiration in plant cells. In Arabidopsis thaliana, submergence induces the transcription of autophagy-related (ATG) genes and the formation of autophagosomes. Consistent with this, the autophagy-defective (atg) mutants are hypersensitive to submergence stress and treatment with ethanol, the end product of anaerobic respiration. Upon submergence, the atg mutants have increased levels of transcripts of anaerobic respiration genes (alcohol dehydrogenase 1, ADH1 and pyruvate decarboxylase 1, PDC1), but reduced levels of transcripts of other hypoxia- and ethylene-responsive genes. Both submergence and ethanol treatments induce the accumulation of reactive oxygen species (ROS) in the rosettes of atg mutants more than in the wild type. Moreover, the production of ROS by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidases is necessary for plant tolerance to submergence and ethanol, submergence-induced expression of ADH1 and PDC1, and activation of autophagy. The submergence- and ethanol-sensitive phenotypes in the atg mutants depend on a complete salicylic acid (SA) signaling pathway. Together, our findings demonstrate that submergence-induced autophagy functions in the hypoxia response in Arabidopsis by modulating SA-mediated cellular homeostasis.
Collapse
Affiliation(s)
- Liang Chen
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Bin Liao
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Hua Qi
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Li-Juan Xie
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Li Huang
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Wei-Juan Tan
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Ning Zhai
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Li-Bing Yuan
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Ying Zhou
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Lu-Jun Yu
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Qin-Fang Chen
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Wensheng Shu
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| | - Shi Xiao
- a State Key Laboratory of Biocontrol; Guangdong Provincial Key Laboratory of Plant Resources; Collaborative Innovation Center of Genetics and Development; School of Life Sciences; Sun Yat-sen University ; Guangzhou , China
| |
Collapse
|
102
|
Liu L, Sun T, Xin F, Cui W, Guo J, Hu J. Nerve Growth Factor Protects Against Alcohol-Induced Neurotoxicity in PC12 Cells via PI3K/Akt/mTOR Pathway. Alcohol Alcohol 2016; 52:12-18. [PMID: 27760741 DOI: 10.1093/alcalc/agw077] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/22/2016] [Accepted: 09/22/2016] [Indexed: 01/26/2023] Open
Abstract
AIMS To study whether autophagy participates in the neuroprotective effect of nerve growth factor (NGF) on neurons treated with alcohol. METHODS The autophagy-related markers were used to explore the role of autophagy in PC12 cells exposed to alcohol or pre-incubated with NGF before initiating the treatment with alcohol (100 mM; 6 h). PC12 cells were pre-incubated with 3-methyladenine (3-MA) (10 mM; 1 h) or rapamycin (100 nM; 1 h) before co-incubated with alcohol (100 mM; 6 h) in order to investigate the relationship between apoptosis and autophagy. PC12 cells were pre-incubated with LY294002 (50 μM; 30 min) before co-incubated with NGF and alcohol in order to analyze the protein expression of PI3K/Akt/mTOR pathway via western blotting. RESULT By methylthiazoltetrazolium, western blotting and flow cytometry assays, we found that cell viability decreased in a dose- and time-dependent manner after treatment with alcohol in PC12 cells. As cells were exposed to alcohol, the levels of LC3-II proteins became elevated, likewise, pre-treatment with 3-methyladenine (3-MA, an autophagic inhibitor) or rapamycin (an autophagic inducer) resulted in an increased or decreased percentage of apoptosis in contrast to other alcohol-treated groups, respectively. NGF markedly increased LC3-II production after treatment with alcohol in a dose-dependent manner. Moreover, NGF remarkably attenuated the phosphorylation effect of alcohol exposure on PI3K/Akt/mTOR pathway, which was suppressed by LY294002 (Akt inhibitor). CONCLUSIONS NGF protects against alcohol-induced neurotoxicity via PI3K/Akt/mTOR pathway. SHORT SUMMARY In this study, we chose the PC12 cell line as a neuronal model, and our results demonstrate that nerve growth factor can induce autophagy with the neuroprotective effect and regulatory mechanisms of alcohol-induced autophagy in PC12 cells.
Collapse
Affiliation(s)
- Liping Liu
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang 150001, PR China
| | - Tiedong Sun
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang Street, Nangang District, Harbin, Heilongjiang 150080, PR China
| | - Feng Xin
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang 150001, PR China
| | - Wei Cui
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang Street, Nangang District, Harbin, Heilongjiang 150080, PR China
| | - Jianquan Guo
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang Street, Nangang District, Harbin, Heilongjiang 150080, PR China
| | - Jian Hu
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang 150001, PR China
| |
Collapse
|
103
|
Song DS, Chang UI, Choi S, Jung YD, Han K, Ko SH, Ahn YB, Yang JM. Heavy Alcohol Consumption with Alcoholic Liver Disease Accelerates Sarcopenia in Elderly Korean Males: The Korean National Health and Nutrition Examination Survey 2008-2010. PLoS One 2016; 11:e0163222. [PMID: 27655344 PMCID: PMC5031399 DOI: 10.1371/journal.pone.0163222] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 09/06/2016] [Indexed: 12/23/2022] Open
Abstract
Background and Aim Although a few studies have reported that sarcopenia is associated with alcoholic liver disease (ALD), no studies have investigated this association in a large sample representative of the elderly Korean population. Methods This was a cross-sectional study that used data from the Fourth and Fifth Korean National Health and Nutrition Examination Surveys (KNHANES) on subjects aged 65 years and older. Sarcopenia was defined as a skeletal muscle index (SMI) more than 1 SD below the gender-specific mean for young adults; SMI was calculated as the appendicular muscle mass divided by height squared (ASM/Ht2). Heavy alcohol consumption was defined as consuming at least 210 g/week, and elevated liver enzymes were defined as alanine aminotransferase levels of at least 32 U/L or aspartate aminotransferase levels of at least 34 U/L. ALD was defined as heavy alcohol consumption and elevated liver enzymes. Results The mean age of the 1,151 elderly males was 71.6 ± 0.2 years, and the prevalence of heavy alcohol consumption was 11.8% (136 subjects). SMI did not differ between the non-heavy and heavy alcohol consumer groups (7.1 ± 0.0 kg/m2 vs. 7.3 ± 0.1 kg/m2, respectively, P = 0.145). However, after stratifying by the presence of liver disease and heavy alcohol consumption and adjusting for other confounders in the multivariate logistic regression, SMI was significantly lower among heavy alcohol consumers with ALD (all P < 0.05). Additionally, two-way ANOVA showed a significant interaction between heavy alcohol consumption and liver disease (P = 0.011). Conclusion Sarcopenia was accelerated in the elderly male ALD group, with a significant interaction between alcohol consumption and liver disease.
Collapse
Affiliation(s)
- Do Seon Song
- Division of Hepatology, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - U Im Chang
- Division of Hepatology, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Sooa Choi
- Division of Hepatology, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Yun Duk Jung
- Division of Hepatology, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Kyungdo Han
- Department of Biostatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Hyun Ko
- Division of Endocrinology and Metabolism, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Yu-Bae Ahn
- Division of Endocrinology and Metabolism, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
- * E-mail: (YBA); (JMY)
| | - Jin Mo Yang
- Division of Hepatology, Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
- * E-mail: (YBA); (JMY)
| |
Collapse
|
104
|
Stern RA, Dasarathy S, Mozdziak PE. Ammonia elicits a different myogenic response in avian and murine myotubes. In Vitro Cell Dev Biol Anim 2016; 53:99-110. [PMID: 27573411 DOI: 10.1007/s11626-016-0088-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/11/2016] [Indexed: 12/27/2022]
Abstract
Increased myostatin expression, resulting in muscle loss, has been associated with hyperammonemia in mammalian models of cirrhosis. However, there is evidence that hyperammonemia in avian embryos results in a reduction of myostatin expression, suggesting a proliferative myogenic environment. The present in vitro study examines species differences in myotube and liver cell response to ammonia using avian and murine-derived cells. Primary myoblasts and liver cells were isolated from embryonic day 15 and 17 chick embryos to be compared with mouse myoblasts (C2C12) and liver (AML12) cells. Cells were exposed to varying concentrations of ammonium acetate (AA; 2.5, 5, or 10 mM) to determine the effects of ammonia on the cells. Relative expression of myostatin mRNA, determined by quantitative real-time PCR, was significantly increased in AA (10 mM) treated C2C12 myotubes compared to both ages of chick embryonic myotube cultures after 48 h (P < 0.02). Western blot analysis of myostatin protein confirmed an increase in myostatin expression in AA-treated C2C12 myotubes compared to the sodium acetate (SA) controls, while myostatin expression was decreased in the chick embryonic myotube cultures when treated with AA. Myotube diameter was significantly decreased in AA-treated C2C12 myotubes compared to controls, while avian myotube diameter increased with AA treatment (P < 0.001). There were no significant differences between avian and murine liver cell viability, assessed using 2', 7'- bis-(2-carboxyethyl)-5-(and-6-)-carboxyfluorescein, acetoxymethyl ester, when treated with AA. However, after 24 h, AA-treated avian myotubes showed a significant increase in cell viability compared to the C2C12 myotubes (P < 0.05). Overall, it appears that there is a positive myogenic response to hyperammonemia in avian myotubes compared to murine myotubes, which supports a proliferative myogenic environment.
Collapse
Affiliation(s)
- Rachel A Stern
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, 27695-7608, USA.
| | - Srinivasan Dasarathy
- Department of Pathobiology, Lerner Research Institute, and Department of Gastroenterology, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Paul E Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, 27695-7608, USA
| |
Collapse
|
105
|
Dasarathy S. Nutrition and Alcoholic Liver Disease: Effects of Alcoholism on Nutrition, Effects of Nutrition on Alcoholic Liver Disease, and Nutritional Therapies for Alcoholic Liver Disease. Clin Liver Dis 2016; 20:535-50. [PMID: 27373615 PMCID: PMC4934388 DOI: 10.1016/j.cld.2016.02.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malnutrition is the most frequent and nearly universal consequence in alcoholic liver disease (ALD) that adversely affects clinical outcomes. Sarcopenia or skeletal muscle loss is the major component of malnutrition in liver disease. There are no effective therapies to prevent or reverse sarcopenia in ALD because the mechanisms are not well understood. Consequences of liver disease including hyperammonemia, hormonal perturbations, endotoxemia and cytokine abnormalities as well as the direct effects of alcohol and its metabolites contribute to sarcopenia in ALD. This article focuses on the prevalence, methods to quantify malnutrition, specifically sarcopenia and potential therapies including novel molecular targeted treatments.
Collapse
Affiliation(s)
- Srinivasan Dasarathy
- Departments of Gastroenterology, Hepatology and Pathobiology, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
106
|
Steffl M, Bohannon RW, Petr M, Kohlikova E, Holmerova I. Alcohol consumption as a risk factor for sarcopenia - a meta-analysis. BMC Geriatr 2016; 16:99. [PMID: 27170042 PMCID: PMC4865026 DOI: 10.1186/s12877-016-0270-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 04/29/2016] [Indexed: 12/22/2022] Open
Abstract
Background Sarcopenia, a loss of muscle strength and mass, has serious implications for older adults. Some risk factors for sarcopenia are well established. The role of other factors such as alcohol consumption is less certain. The main aim of this study was to explore the relationship between sarcopenia and alcohol consumption in people over 65 years old. Methods Four electronic databases were searched to identify potentially relevant papers. Demographics and information on sarcopenia and alcohol consumption were extracted from relevant papers. The relationship between sarcopenia and alcohol consumption was described using odds ratios (ORs). Results Of 214 papers identified as potentially relevant, 13 were ultimately included in the meta-analyses. The papers provided data from 13,155 participants. The OR (95 % CI) for sarcopenia among alcohol drinkers was 0.67 (0.54–0.83) for males, 0.89 (0.73–1.08) for females, and 0.77 (0.67–0.88) for the overall population. Conclusions The results of this meta-analysis do not support alcohol consumption as a risk factor for sarcopenia.
Collapse
Affiliation(s)
- Michal Steffl
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, Charles University Prague, Prague, Czech Republic.
| | - Richard W Bohannon
- Department of Physical Therapy, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC, USA
| | - Miroslav Petr
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, Charles University Prague, Prague, Czech Republic
| | - Eva Kohlikova
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, Charles University Prague, Prague, Czech Republic
| | - Iva Holmerova
- Centre of Gerontology, Faculty of Humanities, Charles University Prague, Prague, Czech Republic
| |
Collapse
|
107
|
Abstract
PURPOSE OF REVIEW Sarcopenia or loss of skeletal muscle mass is the major component of malnutrition and occurs in the majority of patients with liver disease. Lower muscle contractile function also contributes to the adverse consequences of sarcopenia. There are no effective therapies to prevent or reverse sarcopenia in liver disease. This review will discuss the advances in diagnosis, pathogenesis, and treatment options for sarcopenia in liver disease. RECENT FINDINGS Sarcopenia increases mortality and risk of development of other complications of cirrhosis, and worsens postliver transplant outcomes while quality of life is decreased. Unlike other complications of cirrhosis that reverse after liver transplantation, sarcopenia may not improve and actually worsens. Impaired skeletal muscle protein synthesis and increased proteolysis via autophagy contribute to sarcopenia. Hyperammonemia is the best-studied mediator of the liver-muscle axis. Molecular studies show increased expression of myostatin whereas metabolic studies show impaired mitochondrial function and tricarboxylic acid cycle intermediates because of cataplerosis of α-ketoglutarate. Impaired skeletal muscle pyruvate and fatty acid oxidation during hyperammonemia suggest amino acids are diverted to acetyl CoA and potentially aggravate hyperammonemia. Nutritional supplementation is of limited or no benefit and suggests that cirrhosis is a state of anabolic resistance. Exercise may be beneficial but whether it overcomes anabolic resistance is not known. SUMMARY The high clinical significance of sarcopenia is well established. Current approaches to nutritional supplementation have not been effective in reversing sarcopenia because of anabolic resistance. Myostatin antagonists, specific amino acid supplementation, mitochondrial protection, and combination endurance-resistance exercise are potential future therapeutic options.
Collapse
|
108
|
Sinclair M, Gow PJ, Grossmann M, Angus PW. Review article: sarcopenia in cirrhosis--aetiology, implications and potential therapeutic interventions. Aliment Pharmacol Ther 2016; 43:765-77. [PMID: 26847265 DOI: 10.1111/apt.13549] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/12/2015] [Accepted: 01/14/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sarcopenia (loss of muscle mass) is common in cirrhosis and is associated with poor outcomes. Current teaching recommends the use of protein supplementation and exercise, however, this fails to address many other factors which contribute to muscle loss in this setting. AIMS To summarise existing knowledge regarding the aetiology of sarcopenia in cirrhosis, diagnostic modalities and the clinical significance of this condition. In addition to discuss recent research findings that may allow the development of more effective treatments. METHODS We conducted a Medline and PubMed search using the search terms 'sarcopenia', 'muscle', 'body composition', 'cirrhosis', 'liver' and 'malnutrition' from inception to October 2015. RESULTS Cirrhotic patients with sarcopenia have reduced survival, experience increased rates of infection and have worse outcomes following liver transplantation. The aetiology of this condition is more complex than simple protein and calorie malnutrition. Cirrhosis also results in depleted glycogen stores and metabolic alterations that cause excessive protein catabolism, increased activation of the ubiquitin-proteasome pathway and inappropriate muscle autophagy. Satellite cell differentiation and proliferation is also reduced due to a combination of elevated myostatin levels, reduced IGF-1 and hypogonadism. Although there is some evidence supporting the use of late evening snacks, branched chain amino acid supplementation and high protein/high calorie diets, well designed clinical trials addressing the effects of treatment on body composition in cirrhosis are lacking. CONCLUSION Sarcopenia in cirrhosis has a complex pathogenesis and simple dietary interventions are insufficient. Improved understanding of the multiple mechanisms involved should allow the development of more effective therapies, which target the specific underlying metabolic derangements.
Collapse
Affiliation(s)
- M Sinclair
- Liver Transplant Unit, Austin Hospital, Melbourne, Vic., Australia.,Department of Medicine, The University of Melbourne, Melbourne, Vic., Australia
| | - P J Gow
- Liver Transplant Unit, Austin Hospital, Melbourne, Vic., Australia.,Department of Medicine, The University of Melbourne, Melbourne, Vic., Australia
| | - M Grossmann
- Endocrinology Unit, Austin Hospital, Melbourne, Vic., Australia.,Department of Medicine, The University of Melbourne, Melbourne, Vic., Australia
| | - P W Angus
- Liver Transplant Unit, Austin Hospital, Melbourne, Vic., Australia.,Department of Medicine, The University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
109
|
Tanaka K, Whelan KA, Chandramouleeswaran PM, Kagawa S, Rustgi SL, Noguchi C, Guha M, Srinivasan S, Amanuma Y, Ohashi S, Muto M, Klein-Szanto AJ, Noguchi E, Avadhani NG, Nakagawa H. ALDH2 modulates autophagy flux to regulate acetaldehyde-mediated toxicity thresholds. Am J Cancer Res 2016; 6:781-96. [PMID: 27186430 PMCID: PMC4859883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 01/31/2016] [Indexed: 02/15/2023] Open
Abstract
A polymorphic mutation in the acetaldehyde dehydrogenase 2 (ALDH2) gene has been epidemiologically linked to the high susceptibility to esophageal carcinogenesis for individuals with alcohol use disorders. Mice subjected to alcohol drinking show increased oxidative stress and DNA adduct formation in esophageal epithelia where Aldh2 loss augments alcohol-induced genotoxic effects; however, it remains elusive as to how esophageal epithelial cells with dysfunctional Aldh2 cope with oxidative stress related to alcohol metabolism. Here, we investigated the role of autophagy in murine esophageal epithelial cells (keratinocytes) exposed to ethanol and acetaldehyde. We find that ethanol and acetaldehyde trigger oxidative stress via mitochondrial superoxide in esophageal keratinocytes. Aldh2-deficient cells appeared to be highly susceptible to ethanol- or acetaldehyde-mediated toxicity. Alcohol dehydrogenase-mediated acetaldehyde production was implicated in ethanol-induced cell injury in Aldh2 deficient cells as ethanol-induced oxidative stress and cell death was partially inhibited by 4-methylpyrazole. Acetaldehyde activated autophagy flux in esophageal keratinocytes where Aldh2 deficiency increased dependence on autophagy to cope with ethanol-induced acetaldehyde-mediated oxidative stress. Pharmacological inhibition of autophagy flux by chloroquine stabilized p62/SQSTM1, and increased basal and acetaldehyde-mediate oxidative stress in Aldh2 deficient cells as documented in monolayer culture as well as single-cell derived three-dimensional esophageal organoids, recapitulating a physiological esophageal epithelial proliferation-differentiation gradient. Our innovative approach indicates, for the first time, that autophagy may provide cytoprotection to esophageal epithelial cells responding to oxidative stress that is induced by ethanol and its major metabolite acetaldehyde. Defining autophagymediated cytoprotection against alcohol-induced genotoxicity in the context of Aldh2 deficiency, our study provides mechanistic insights into the tumor suppressor functions of ALDH2 and autophagy in alcohol-related esophageal carcinogenesis.
Collapse
Affiliation(s)
- Koji Tanaka
- Gastroenterology Division, Department of Medicine, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA,University of Pennsylvania Abramson Cancer CenterPhiladelphia, PA, USA
| | - Kelly A Whelan
- Gastroenterology Division, Department of Medicine, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA,University of Pennsylvania Abramson Cancer CenterPhiladelphia, PA, USA
| | - Prasanna M Chandramouleeswaran
- Gastroenterology Division, Department of Medicine, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA,University of Pennsylvania Abramson Cancer CenterPhiladelphia, PA, USA
| | - Shingo Kagawa
- Gastroenterology Division, Department of Medicine, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA,University of Pennsylvania Abramson Cancer CenterPhiladelphia, PA, USA
| | - Sabrina L Rustgi
- Gastroenterology Division, Department of Medicine, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA,University of Pennsylvania Abramson Cancer CenterPhiladelphia, PA, USA
| | - Chiaki Noguchi
- Department of Biochemistry and Molecular Biology, Drexel University College of MedicinePhiladelphia, PA, USA
| | - Manti Guha
- Department of Animal Biology, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of PennsylvaniaPhiladelphia, PA, USA
| | - Satish Srinivasan
- Department of Animal Biology, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of PennsylvaniaPhiladelphia, PA, USA
| | - Yusuke Amanuma
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto UniversityKyoto, Japan
| | - Shinya Ohashi
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto UniversityKyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto UniversityKyoto, Japan
| | - Andres J Klein-Szanto
- Histopathology Facility and Cancer Biology Program, Fox Chase Cancer CenterPhiladelphia, PA, USA
| | - Eishi Noguchi
- Department of Biochemistry and Molecular Biology, Drexel University College of MedicinePhiladelphia, PA, USA
| | - Narayan G Avadhani
- Department of Animal Biology, Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of PennsylvaniaPhiladelphia, PA, USA
| | - Hiroshi Nakagawa
- Gastroenterology Division, Department of Medicine, University of Pennsylvania Perelman School of MedicinePhiladelphia, PA, USA,University of Pennsylvania Abramson Cancer CenterPhiladelphia, PA, USA
| |
Collapse
|
110
|
Lesmana R, Sinha RA, Singh BK, Zhou J, Ohba K, Wu Y, Yau WWY, Bay BH, Yen PM. Thyroid Hormone Stimulation of Autophagy Is Essential for Mitochondrial Biogenesis and Activity in Skeletal Muscle. Endocrinology 2016; 157:23-38. [PMID: 26562261 DOI: 10.1210/en.2015-1632] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thyroid hormone (TH) and autophagy share similar functions in regulating skeletal muscle growth, regeneration, and differentiation. Although TH recently has been shown to increase autophagy in liver, the regulation and role of autophagy by this hormone in skeletal muscle is not known. Here, using both in vitro and in vivo models, we demonstrated that TH induces autophagy in a dose- and time-dependent manner in skeletal muscle. TH induction of autophagy involved reactive oxygen species (ROS) stimulation of 5'adenosine monophosphate-activated protein kinase (AMPK)-Mammalian target of rapamycin (mTOR)-Unc-51-like kinase 1 (Ulk1) signaling. TH also increased mRNA and protein expression of key autophagy genes, microtubule-associated protein light chain 3 (LC3), Sequestosome 1 (p62), and Ulk1, as well as genes that modulated autophagy and Forkhead box O (FOXO) 1/3a. TH increased mitochondrial protein synthesis and number as well as basal mitochondrial O2 consumption, ATP turnover, and maximal respiratory capacity. Surprisingly, mitochondrial activity and biogenesis were blunted when autophagy was blocked in muscle cells by Autophagy-related gene (Atg)5 short hairpin RNA (shRNA). Induction of ROS and 5'adenosine monophosphate-activated protein kinase (AMPK) by TH played a significant role in the up-regulation of Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A), the key regulator of mitochondrial synthesis. In summary, our findings showed that TH-mediated autophagy was essential for stimulation of mitochondrial biogenesis and activity in skeletal muscle. Moreover, autophagy and mitochondrial biogenesis were coupled in skeletal muscle via TH induction of mitochondrial activity and ROS generation.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/chemistry
- AMP-Activated Protein Kinases/metabolism
- Animals
- Autophagy/drug effects
- Autophagy-Related Protein 5
- Autophagy-Related Protein-1 Homolog
- Cell Line
- Gene Expression Regulation/drug effects
- Kinetics
- Male
- Mice, Inbred C57BL
- Microtubule-Associated Proteins/antagonists & inhibitors
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/metabolism
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/ultrastructure
- Mitochondrial Dynamics/drug effects
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- Myoblasts, Skeletal/cytology
- Myoblasts, Skeletal/drug effects
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/ultrastructure
- Oxygen Consumption/drug effects
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
- Protein Serine-Threonine Kinases/chemistry
- Protein Serine-Threonine Kinases/metabolism
- RNA Interference
- Reactive Oxygen Species/agonists
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Thyroxine/metabolism
- Thyroxine/pharmacology
- Transcription Factors/agonists
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Triiodothyronine/metabolism
- Triiodothyronine/pharmacology
Collapse
Affiliation(s)
- Ronny Lesmana
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Rohit A Sinha
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Brijesh K Singh
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Jin Zhou
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Kenji Ohba
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Yajun Wu
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Winifred W Y Yau
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Boon-Huat Bay
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Paul M Yen
- Laboratory of Hormonal Regulation (R.L., R.A.S., B.K.S., J.Z., K.O., W.WY.Y., P.M.Y.), Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Department of Physiology (R.L.), Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Anatomy (Y.W., B.-H.B.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599; and Duke Molecular Physiology Institute and Department of Medicine (P.M.Y.), Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
111
|
Abstract
Excessive ethanol exposure is detrimental to the brain. The developing brain is particularly vulnerable to ethanol such that prenatal ethanol exposure causes fetal alcohol spectrum disorders (FASD). Neuronal loss in the brain is the most devastating consequence and is associated with mental retardation and other behavioral deficits observed in FASD. Since alcohol consumption during pregnancy has not declined, it is imperative to elucidate the underlying mechanisms and develop effective therapeutic strategies. One cellular mechanism that acts as a protective response for the central nervous system (CNS) is autophagy. Autophagy regulates lysosomal turnover of organelles and proteins within cells, and is involved in cell differentiation, survival, metabolism, and immunity. We have recently shown that ethanol activates autophagy in the developing brain. The autophagic preconditioning alleviates ethanol-induced neuron apoptosis, whereas inhibition of autophagy potentiates ethanol-stimulated reactive oxygen species (ROS) and exacerbates ethanol-induced neuroapoptosis. The expression of genes encoding proteins required for autophagy in the CNS is developmentally regulated; their levels are much lower during an ethanol-sensitive period than during an ethanol-resistant period. Ethanol may stimulate autophagy through multiple mechanisms; these include induction of oxidative stress and endoplasmic reticulum stress, modulation of MTOR and AMPK signaling, alterations in BCL2 family proteins, and disruption of intracellular calcium (Ca2+) homeostasis. This review discusses the most recent evidence regarding the involvement of autophagy in ethanol-mediated neurotoxicity as well as the potential therapeutic approach of targeting autophagic pathways.
Collapse
Key Words
- AD, Alzheimer disease
- ALS, autophagy-lysosome system
- AMPK, adenosine 5′-monophosphate-activated protein kinase;
- ATG, autophagy-related
- CNS, central nervous system
- ER, endoplasmic reticulum
- FASD, fetal alcohol spectrum disorders
- FOXO3, forkhead box O3
- GSK3B, glycogen synthase kinase 3 β
- HD, Huntington disease, HNSCs, hippocampal neural stem cells
- LC3, microtubule-associated protein 1 light chain 3
- MTOR, mechanistic target of rapamycin (serine/threonine kinase)
- MTORC1, MTOR complex 1
- NFE2L2, nuclear factor, erythroid 2-like 2
- NOX, NADPH oxidase
- PD, Parkinson disease
- PI3K, class I phosphoinositide 3-kinase
- ROS, reactive oxygen species
- SQSTM1/p62, sequestosome 1
- TSC1/2, tuberous sclerosis 1/ 2
- UPR, unfolded protein response
- alcohol
- alcoholism
- development
- fetal alcohol spectrum disorders
- neurodegeneration
- oxidative stress
- protein degradation
Collapse
Affiliation(s)
- Jia Luo
- a Department of Pharmacology and Nutritional Sciences ; University of Kentucky College of Medicine ; Lexington , KY USA
| |
Collapse
|
112
|
Abstract
High mobility group box 1 (HMGB1) plays a key role in human health and disease. Currently, three different labs in the USA use Cre-lox technology to create mice with a hepatocyte-specific Hmgb1 deletion (HMGB1-HC-KO mice) by backcrossing Hmgb1 (flox)/(flox) mice to Alb-Cre mice. This mouse strain has a different phenotype following exposure to several stressors.
Collapse
Affiliation(s)
- Xiaofang Sun
- a The Third Affiliated Hospital of Guangzhou Medical University ; Guangzhou , Guangdong , China
| | | |
Collapse
|
113
|
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: 19] [Impact Index Per Article: 2.1] [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
|
114
|
Tsien C, Davuluri G, Singh D, Allawy A, Ten Have GAM, Thapaliya S, Schulze JM, Barnes D, McCullough AJ, Engelen MP, Deutz NE, Dasarathy S. Metabolic and molecular responses to leucine-enriched branched chain amino acid supplementation in the skeletal muscle of alcoholic cirrhosis. Hepatology 2015; 61:2018-29. [PMID: 25613922 PMCID: PMC4441611 DOI: 10.1002/hep.27717] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/22/2014] [Accepted: 01/20/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED Skeletal muscle loss (sarcopenia) is a major clinical complication in alcoholic cirrhosis with no effective therapy. Skeletal muscle autophagic proteolysis and myostatin expression (inhibitor of protein synthesis) are increased in cirrhosis and believed to contribute to anabolic resistance. A prospective study was performed to determine the mechanisms of sarcopenia in alcoholic cirrhosis and potential reversal by leucine. In six well-compensated, stable, alcoholic patients with cirrhosis and eight controls, serial vastus lateralis muscle biopsies were obtained before and 7 hours after a single oral branched chain amino acid mixture enriched with leucine (BCAA/LEU). Primed-constant infusion of l-[ring-(2) H5 ]-phenylalanine was used to quantify whole-body protein breakdown and muscle protein fractional synthesis rate using liquid chromatography/mass spectrometry. Muscle expression of myostatin, mammalian target of rapamycin (mTOR) targets, autophagy markers, protein ubiquitination, and the intracellular amino acid deficiency sensor general control of nutrition 2 were quantified by immunoblots and the leucine exchanger (SLC7A5) and glutamine transporter (SLC38A2), by real-time polymerase chain reaction. Following oral administration, plasma BCAA concentrations showed a similar increase in patients with cirrhosis and controls. Skeletal muscle fractional synthesis rate was 9.63 ± 0.36%/hour in controls and 9.05 ± 0.68%/hour in patients with cirrhosis (P = 0.54). Elevated whole-body protein breakdown in patients with cirrhosis was reduced with BCAA/LEU (P = 0.01). Fasting skeletal muscle molecular markers showed increased myostatin expression, impaired mTOR signaling, and increased autophagy in patients with cirrhosis compared to controls (P < 0.01). The BCAA/LEU supplement did not alter myostatin expression, but mTOR signaling, autophagy measures, and general control of nutrition 2 activation were consistently reversed in cirrhotic muscle (P < 0.01). Expression of SLC7A5 was higher in the basal state in patients with cirrhosis than controls (P < 0.05) but increased with BCAA/LEU only in controls (P < 0.001). CONCLUSIONS Impaired mTOR1 signaling and increased autophagy in skeletal muscle of patients with alcoholic cirrhosis is acutely reversed by BCAA/LEU.
Collapse
Affiliation(s)
- Cynthia Tsien
- Departments of Gastroenterology, Toronto General Hospital, Toronto, Canada
| | | | - Dharmvir Singh
- Departments of Gastroenterology, Hepatology, Cleveland Clinic, Cleveland Ohio
| | - Allawy Allawy
- Department of Pathobiology, Cleveland Clinic, Cleveland Ohio
| | | | - Samjhana Thapaliya
- Departments of Gastroenterology, Hepatology, Cleveland Clinic, Cleveland Ohio
| | - John M Schulze
- Molecular Structure Facility, University of California, Davis, California
| | - David Barnes
- Departments of Gastroenterology, Hepatology, Cleveland Clinic, Cleveland Ohio
| | | | | | | | - Srinivasan Dasarathy
- Department of Pathobiology, Cleveland Clinic, Cleveland Ohio,Departments of Gastroenterology, Hepatology, Cleveland Clinic, Cleveland Ohio
| |
Collapse
|
115
|
Steiner JL, Lang CH. Dysregulation of skeletal muscle protein metabolism by alcohol. Am J Physiol Endocrinol Metab 2015; 308:E699-712. [PMID: 25759394 PMCID: PMC4420901 DOI: 10.1152/ajpendo.00006.2015] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/03/2015] [Indexed: 12/12/2022]
Abstract
Alcohol abuse, either by acute intoxication or prolonged excessive consumption, leads to pathological changes in many organs and tissues including skeletal muscle. As muscle protein serves not only a contractile function but also as a metabolic reserve for amino acids, which are used to support the energy needs of other tissues, its content is tightly regulated and dynamic. This review focuses on the etiology by which alcohol perturbs skeletal muscle protein balance and thereby over time produces muscle wasting and weakness. The preponderance of data suggest that alcohol primarily impairs global protein synthesis, under basal conditions as well as in response to several anabolic stimuli including growth factors, nutrients, and muscle contraction. This inhibitory effect of alcohol is mediated, at least in part, by a reduction in mTOR kinase activity via a mechanism that remains poorly defined but likely involves altered protein-protein interactions within mTOR complex 1. Furthermore, alcohol can exacerbate the decrement in mTOR and/or muscle protein synthesis present in other catabolic states. In contrast, alcohol-induced changes in muscle protein degradation, either global or via specific modulation of the ubiquitin-proteasome or autophagy pathways, are relatively inconsistent and may be model dependent. Herein, changes produced by acute intoxication versus chronic ingestion are contrasted in relation to skeletal muscle metabolism, and limitations as well as opportunities for future research are discussed. As the proportion of more economically developed countries ages and chronic illness becomes more prevalent, a better understanding of the etiology of biomedical consequences of alcohol use disorders is warranted.
Collapse
Affiliation(s)
- Jennifer L Steiner
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
116
|
Steiner JL, Gordon BS, Lang CH. Moderate alcohol consumption does not impair overload-induced muscle hypertrophy and protein synthesis. Physiol Rep 2015; 3:3/3/e12333. [PMID: 25780086 PMCID: PMC4393167 DOI: 10.14814/phy2.12333] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Chronic alcohol consumption leads to muscle weakness and atrophy in part by suppressing protein synthesis and mTORC1-mediated signaling. However, it is unknown whether moderate alcohol consumption also prevents overload-induced muscle growth and related anabolic signaling. Hypertrophy of the plantaris muscle was induced by removal of a section of the gastrocnemius and soleus muscles from one leg of C57BL/6 adult male mice while the contralateral leg remained intact as the sham control. A nutritionally complete alcohol-containing liquid diet (EtOH) or isocaloric, alcohol-free liquid diet (Con) was provided for 14 days post-surgery. EtOH intake was increased progressively (day 1-5) before being maintained at ~20 g/day/kg BW. The plantaris muscle from the sham and OL leg was removed after 14 days at which time there was no difference in body weight between Con and EtOH-fed mice. OL increased muscle weight (90%) and protein synthesis (125%) in both Con and EtOH mice. The overload-induced increase in mTOR (Ser2448), 4E-BP1 (Thr37/46), S6K1 (Thr389), rpS6 (Ser240/244), and eEF2 (Thr56) were comparable in muscle from Con and EtOH mice. Modulation of signaling upstream of mTORC1 including REDD1 protein expression, Akt (Thr308), PRAS40 (Thr246), and ERK (Thr202/Tyr204) also did not differ between Con and EtOH mice. Markers of autophagy (ULK1, p62, and LC3) suggested inhibition of autophagy with overload and activation with alcohol feeding. These data show that moderate alcohol consumption does not impair muscle growth, and therefore imply that resistance exercise may be an effective therapeutic modality for alcoholic-related muscle disease.
Collapse
Affiliation(s)
- Jennifer L Steiner
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Bradley S Gordon
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
117
|
Autophagy in alcohol-induced multiorgan injury: mechanisms and potential therapeutic targets. BIOMED RESEARCH INTERNATIONAL 2014; 2014:498491. [PMID: 25140315 PMCID: PMC4124834 DOI: 10.1155/2014/498491] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 06/29/2014] [Indexed: 12/21/2022]
Abstract
Autophagy is a genetically programmed, evolutionarily conserved intracellular degradation pathway involved in the trafficking of long-lived proteins and cellular organelles to the lysosome for degradation to maintain cellular homeostasis. Alcohol consumption leads to injury in various tissues and organs including liver, pancreas, heart, brain, and muscle. Emerging evidence suggests that autophagy is involved in alcohol-induced tissue injury. Autophagy serves as a cellular protective mechanism against alcohol-induced tissue injury in most tissues but could be detrimental in heart and muscle. This review summarizes current knowledge about the role of autophagy in alcohol-induced injury in different tissues/organs and its potential molecular mechanisms as well as possible therapeutic targets based on modulation of autophagy.
Collapse
|
118
|
Dasarathy S. Treatment to improve nutrition and functional capacity evaluation in liver transplant candidates. ACTA ACUST UNITED AC 2014; 12:242-55. [PMID: 24691782 DOI: 10.1007/s11938-014-0016-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OPINION STATEMENT Liver transplantation is the definitive therapy for cirrhosis, and malnutrition is the most frequent complication in these patients. Sarcopenia or loss of muscle mass is the major component of malnutrition in cirrhotics and adversely affects their outcome. In addition to the metabolic consequences, functional consequences of sarcopenia include reduced muscle strength and deconditioning. Despite nearly universal occurrence of sarcopenia and its attendant complications, there are no established therapies to prevent or reverse the same. Major reasons for this deficiency include the lack of established standardized definitions or measures to quantify muscle mass, as well as paucity of mechanistic studies or identified molecular targets to develop specific therapeutic interventions. Anthropometric evaluation, bioelectrical impedance analysis, and DEXA scans are relatively imprecise measures of muscle mass, and recent data on imaging measures to determine muscle mass accurately are likely to allow well-defined outcome responses to treatments. Resurgence of interest in the mechanisms of muscle loss in liver disease has been directly related to the rapid advances in the field of muscle biology. Metabolic tracer studies on whole body kinetics have been complemented by direct studies on the skeletal muscle of cirrhotics. Hypermetabolism and anabolic resistance contribute to sarcopenia. Reduced protein synthesis and increased autophagy have been reported in cirrhotic skeletal muscle, while the contribution of the ubiquitin-proteasome pathway is controversial. Increased plasma concentration and skeletal muscle expression of myostatin, a TGFβ superfamily member that causes reduction in muscle mass, have been reported in cirrhosis. Hyperammonemia and TNFα have been reported to increase myostatin expression and may be responsible for sarcopenia in cirrhosis. Nutriceutical interventions with leucine enriched amino acid mixtures, myostatin antagonists and physical activity hold promise as measures to reverse sarcopenia. There is even less data on muscle function and deconditioning in cirrhosis and studies in this area are urgently needed. Even though macronutrient replacement is a major therapeutic goal, micronutrient supplementation, specifically vitamin D, is expected to improve outcomes.
Collapse
Affiliation(s)
- Srinivasan Dasarathy
- Lerner Research Institute, NE4 208, 9500 Euclid Avenue, Cleveland, OH, 44195, USA,
| |
Collapse
|