1
|
Deminice R, Cella P, Borsari A, Padilha C, de Oliveira V. Angiotensin II Type 1 Receptor Blocker Usage Prevents Oxidative Stress and Muscle Dysfunction in HIV. Fundam Clin Pharmacol 2025; 39:e70016. [PMID: 40421794 PMCID: PMC12108033 DOI: 10.1111/fcp.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/02/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND We aimed to elucidate the role of Angiotensin II type 1 receptor (AT1R) blocker usage in muscle wasting and dysfunction related to HIV. RESEARCH DESIGN AND METHODS Appendicular skeletal muscle mass, higher and lower limb strength, and physical fitness were determined in people living with HIV (PWH) using AT1R blockers users (n = 33), angiotensin-converting enzyme (ACE) inhibitors (n = 28), or not using antihypertensive drugs (n = 33). Groups had similar age, sex, race, BMI, and time of HIV infection. Muscle biopsies were performed to determine the abundance of AT1R, the relative abundance of selected proteins related to proteolysis, antioxidant enzymes, and oxidative stress. Plasma angiotensin II, IL-6, and TNF-alpha were also determined. RESULTS PWH using AT1R blocker presented higher strength, physical fitness, and muscle mass than PWH using ACE inhibitors or not using antihypertensive drugs. Although both PWH using AT1R blockers and ACE inhibitors presented reduced angiotensin II plasma levels, only PWH using AT1R blockers presented lower skeletal muscle AT1R activation, lower plasma oxidative stress markers, lower skeletal muscle oxidative stress (4-HNE), and proteolysis markers (Atrogin-1, Murf-1). CONCLUSION AT1R blocker usage protects against oxidative stress and activated proteolysis, contributing to the prevention of muscle wasting and dysfunction among PWH.
Collapse
Affiliation(s)
- Rafael Deminice
- Physical Education and Sports InstituteState University of LondrinaLondrinaBrazil
| | - Paola Sanches Cella
- Physical Education and Sports InstituteState University of LondrinaLondrinaBrazil
| | - Ana Lúcia Borsari
- Physical Education and Sports InstituteState University of LondrinaLondrinaBrazil
| | - Camila S. Padilha
- Physical Education and Sports InstituteState University of LondrinaLondrinaBrazil
- Biology of Ageing Laboratory, Centre for Healthy Ageing, Centenary Institute of Cancer Medicine and Cell BiologyRoyal Prince Alfred HospitalSidneyNSWAustralia
- Faculty of Medicine and Health, Charles Perkins CentreThe University of SydneySydneyNSWAustralia
- Faculty of Health, School of Sport, Exercise and Rehabilitation SciencesUniversity of Technology SydneyUltimoNSWAustralia
| | - Vitor Hugo Fernando de Oliveira
- Physical Education and Sports InstituteState University of LondrinaLondrinaBrazil
- Department of Child, Family and Population Health NursingUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
2
|
Afsar B, Afsar RE, Caliskan Y, Lentine KL, Edwards JC. Renin angiotensin system-induced muscle wasting: putative mechanisms and implications for clinicians. Mol Cell Biochem 2025; 480:1935-1949. [PMID: 38811433 PMCID: PMC11961475 DOI: 10.1007/s11010-024-05043-8] [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: 02/25/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Renin angiotensin system (RAS) alters various mechanisms related to muscle wasting. The RAS system consists of classical and non-classical pathways, which mostly function differently. Classical RAS pathway, operates through angiotensin II (AngII) and angiotensin type 1 receptors, is associated with muscle wasting and sarcopenia. On the other hand, the non-classical RAS pathway, which operates through angiotensin 1-7 and Mas receptor, is protective against sarcopenia. The classical RAS pathway might induce muscle wasting by variety of mechanisms. AngII reduces body weight, via reduction in food intake, possibly by decreasing hypothalamic expression of orexin and neuropeptide Y, insulin like growth factor-1 (IGF-1) and mammalian target of rapamycin (mTOR), signaling, AngII increases skeletal muscle proteolysis by forkhead box transcription factors (FOXO), caspase activation and muscle RING-finger protein-1 transcription. Furthermore, AngII infusion in skeletal muscle reduces phospho-Bad (Ser136) expression and induces apoptosis through increased cytochrome c release and DNA fragmentation. Additionally, Renin angiotensin system activation through AT1R and AngII stimulates tumor necrosis factor-α, and interleukin-6 which induces muscle wasting, Last but not least classical RAS pathway, induce oxidative stress, disturb mitochondrial energy metabolism, and muscle satellite cells which all lead to muscle wasting and decrease muscle regeneration. On the contrary, the non-classical RAS pathway functions oppositely to mitigate these mechanisms and protects against muscle wasting. In this review, we summarize the mechanisms of RAS-induced muscle wasting and putative implications for clinical practice. We also emphasize the areas of uncertainties and suggest potential research areas.
Collapse
Affiliation(s)
- Baris Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey.
- Division of Nephrology, School of Medicine, Saint Louis University, St. Louis, MO, USA.
| | - Rengin Elsurer Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey
- Division of Nephrology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Yasar Caliskan
- Division of Nephrology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Krista L Lentine
- Division of Nephrology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - John C Edwards
- Division of Nephrology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| |
Collapse
|
3
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
4
|
Kakar SS, Vemuri V, Ratajczak MZ. Withaferin A Attenuates Muscle Cachexia Induced by Angiotensin II Through Regulating Pathways Activated by Angiotensin II. Cells 2025; 14:244. [PMID: 39996717 PMCID: PMC11853093 DOI: 10.3390/cells14040244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Cachexia is a multifactorial syndrome characterized by severe muscle wasting and is a debilitating condition frequently associated with cancer. Previous studies from our group revealed that withaferin A (WFA), a steroidal lactone, mitigated muscle cachexia induced by ovarian tumors in NSG mice. However, it remains unclear whether WFA's protective effects are direct or secondary to its antitumor properties. We developed a cachectic model through continuous angiotensin II (Ang II) infusion in C57BL/6 mice to address this issue. Ang II infusion resulted in profound muscle atrophy, evidenced by significant reductions in grip strength and in the TA, GA, and GF muscle mass. Molecular analyses indicated elevated expression of inflammatory cytokines (TNFα, IL-6, MIP-2, IL-18, IL-1β), NLRP3 inflammasome, and genes associated with the UPS (MuRF1, MAFBx) and autophagy pathways (Bacl1, LC3B), along with suppression of anti-inflammatory heme oxygenase-1 (HO-1) and myogenic regulators (Pax7, Myod1). Strikingly, WFA treatment reversed these pathological changes, restoring muscle mass, strength, and molecular markers to near-normal levels. These findings demonstrate that WFA exerts direct anti-cachectic effects by targeting key inflammatory and atrophic pathways in skeletal muscle, highlighting its potential as a novel therapeutic agent for cachexia management.
Collapse
Affiliation(s)
- Sham S. Kakar
- Department of Physiology, University of Louisville, Louisville, KY 40202, USA;
- Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA;
| | - Vasa Vemuri
- Department of Physiology, University of Louisville, Louisville, KY 40202, USA;
| | - Mariusz Z. Ratajczak
- Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA;
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
5
|
Takeshita H, Yamamoto K, Mogi M, Rakugi H. Muscle mass, muscle strength and the renin-angiotensin system. Clin Sci (Lond) 2024; 138:1561-1577. [PMID: 39718491 DOI: 10.1042/cs20220501] [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: 01/26/2023] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024]
Abstract
The renin-angiotensin system (RAS) is a classically known circulatory regulatory system. In addition to the previously known multi-organ circulatory form of the RAS, the existence of tissue RASs in individual organs has been well established. Skeletal muscle has also been identified as an organ with a distinct RAS. In recent years, the effects of RAS activation on skeletal muscle have been elucidated from several perspectives: differences in motor function due to genetic polymorphisms of RAS components, skeletal muscle dysfunction under conditions of excessive RAS activation such as heart failure, and the effects of the use of RAS inhibitors on muscle strength. In addition, the concept of the RAS itself has recently been expanded with the discovery of a 'protective arm' of the RAS formed by factors such as angiotensin-converting enzyme 2 and angiotensin 1-7. This has led to a new understanding of the physiological function of the RAS in skeletal muscle. This review summarizes the diverse physiological functions of the RAS in skeletal muscle and considers the potential of future therapeutic strategies targeting the RAS to overcome problems such as sarcopenia and muscle weakness associated with chronic disease.
Collapse
Affiliation(s)
- Hikari Takeshita
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Medical Science Technology, Faculty of Medical Science Technology, Morinomiya University of Medical Sciences, Osaka, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | | |
Collapse
|
6
|
Santos DCD, Alves FHF, Veríssimo LF, Raquel HA, Volpini VL, Marques LADC, Martins-Pinge MC, Fernandes KBP, Andrade KC, Michelini LC, Pelosi GG. Enalapril induces muscle epigenetic changes and contributes to prevent a decline in running capacity in spontaneously hypertensive rats. Arch Gerontol Geriatr 2024; 129:105699. [PMID: 39581157 DOI: 10.1016/j.archger.2024.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
Drugs such as angiotensin-converting enzyme inhibitors and angiotensin receptor blockers can improve muscle function and exercise capacity, as well as preventing, attenuating or reversing age-related losses in muscle mass, however, the exact mechanisms by which these drugs affect muscle cells, are not yet fully elucidated. Moreover, the potential epigenetic alterations induced in skeletal muscle tissue are also largely unexplored. The aim of this study was to evaluate if enalapril or losartan can change the physical performance and epigenetic profile of skeletal muscle in spontaneously hypertensive rats (SHRs). Male SHRs were treated with water, enalapril (10/mg/kg/day) or losartan (10/mg/kg/day) for 28 consecutive days and submitted to progressive testing on a treadmill. Body weight, perigonadal and retroperitoneal fat, mean arterial pressure, heart rate, running distance and global DNA methylation in the gastrocnemius and soleus muscles were evaluated. Enalapril reduced the rate of weight gain, as well as reducing retroperitoneal fat (p < 0.05) and MAP (p < 0.05) and avoiding the decline in running distance when compared to the other groups (p > 0.05), even 7 days after the end of treatment (p > 0.05). Moreover, enalapril increased global DNA methylation in gastrocnemius muscle cells (p < 0.01). No effects were observed in the losartan-treated group. Our data showed that enalapril prevented the decline in physical function in SHR, as well as reduced the rate of weight gain of the animals. In addition, the results showed, alterations in the global DNA methylation of skeletal muscle cells skeletal structures of the gastrocnemius muscle.
Collapse
Affiliation(s)
- Denis Carlos Dos Santos
- Department of Physiological Sciences, Biological Sciences Center, State University of Londrina (UEL), Londrina, Paraná, Brazil
| | - Fernando Henrique Ferrari Alves
- Department of Health Sciences Faculty of Medicine Federal University of Lavras (UFLA), Lavras, Minas Gerais, Brazil; Institute of Science, Technology and Innovation - Federal University of Lavras, São Sebastião do Paraíso, MG, Brazil.
| | - Luiz Fernando Veríssimo
- Department of Physiological Sciences, Biological Sciences Center, State University of Londrina (UEL), Londrina, Paraná, Brazil
| | - Hiviny Ataides Raquel
- Department of Physiological Sciences, Biological Sciences Center, State University of Londrina (UEL), Londrina, Paraná, Brazil
| | - Vinicius Lucca Volpini
- Department of Physiological Sciences, Biological Sciences Center, State University of Londrina (UEL), Londrina, Paraná, Brazil
| | - Leonardo André da Costa Marques
- Department of Physiological Sciences, Biological Sciences Center, State University of Londrina (UEL), Londrina, Paraná, Brazil
| | - Marli Cardoso Martins-Pinge
- Department of Physiological Sciences, Biological Sciences Center, State University of Londrina (UEL), Londrina, Paraná, Brazil
| | - Karen Barros Parron Fernandes
- School of Medicine, Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Londrina, Paraná, Brazil
| | - Karoliny Coelho Andrade
- Department of Health Sciences Faculty of Medicine Federal University of Lavras (UFLA), Lavras, Minas Gerais, Brazil
| | - Lisete Compagno Michelini
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Gislaine Garcia Pelosi
- Department of Physiological Sciences, Biological Sciences Center, State University of Londrina (UEL), Londrina, Paraná, Brazil
| |
Collapse
|
7
|
Proença AB, Alexandre‐Santos B, Giori IG, Alex‐Marques JSF, Machado‐Santos C, Machado M, Magliano DC, da Nobrega ACL, Frantz EDC. Obesity-induced skeletal muscle remodeling: A comparative analysis of exercise training and ACE-inhibitory drug in male mice. Physiol Rep 2024; 12:e16025. [PMID: 38684378 PMCID: PMC11058004 DOI: 10.14814/phy2.16025] [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: 12/01/2023] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity over-activates the classical arm of the renin-angiotensin system (RAS), impairing skeletal muscle remodeling. We aimed to compare the effect of exercise training and enalapril, an angiotensin-converting enzyme inhibitor, on RAS modulation in the skeletal muscle of obese animals. Thus, we divided C57BL/6 mice into two groups: standard chow (SC) and high-fat (HF) diet for 16 weeks. At the eighth week, the HF-fed animals were divided into four subgroups-sedentary (HF), treated with enalapril (HF-E), exercise training protocol (HF-T), and combined interventions (HF-ET). After 8 weeks of treatment, we evaluated body mass and index (BMI), body composition, exercise capacity, muscle morphology, and skeletal muscle molecular markers. All interventions resulted in lower BMI and attenuation of overactivation in the classical arm, while favoring the B2R in the bradykinin receptors profile. This was associated with reduced apoptosis markers in obese skeletal muscles. The HF-T group showed an increase in muscle mass and expression of biosynthesis markers and a reduction in expression of degradation markers and muscle fiber atrophy due to obesity. These findings suggest that the combination intervention did not have a synergistic effect against obesity-induced muscle remodeling. Additionally, the use of enalapril impaired muscle's physiological adaptations to exercise training.
Collapse
Affiliation(s)
- Ana Beatriz Proença
- Laboratory of Exercise Sciences, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
- Research Center on Morphology and Metabolism, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
| | - Beatriz Alexandre‐Santos
- Laboratory of Exercise Sciences, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
- Research Center on Morphology and Metabolism, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
| | - Isabele Gomes Giori
- Laboratory of Exercise Sciences, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
| | - Jaime Silva Filho Alex‐Marques
- Laboratory of Exercise Sciences, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
- Research Center on Morphology and Metabolism, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
| | - Clarice Machado‐Santos
- Laboratory of Teaching and Research in Histology and Compared EmbryologyFluminense Federal UniversityNiteroiRio de JaneiroBrazil
| | - Marcus Machado
- Biomedical Science DepartmentRoss University School of Veterinary MedicineBasseterreSt. Kitts & Nevis
| | - D'Angelo Carlo Magliano
- Research Center on Morphology and Metabolism, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
| | - Antonio Claudio Lucas da Nobrega
- Laboratory of Exercise Sciences, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
- National Institute for Science & Technology—INCT Physical (in)Activity & Exercise, CNPqNiteroiRio de JaneiroBrazil
| | - Eliete Dalla Corte Frantz
- Laboratory of Exercise Sciences, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
- Research Center on Morphology and Metabolism, Biomedical InstituteFluminense Federal UniversityNiteroiRio de JaneiroBrazil
- National Institute for Science & Technology—INCT Physical (in)Activity & Exercise, CNPqNiteroiRio de JaneiroBrazil
| |
Collapse
|
8
|
Hirunsai M, Srikuea R. Differential effects of cholecalciferol and calcitriol on muscle proteolysis and oxidative stress in angiotensin II-induced C2C12 myotube atrophy. Physiol Rep 2024; 12:e16011. [PMID: 38627219 PMCID: PMC11021198 DOI: 10.14814/phy2.16011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/13/2024] [Accepted: 03/29/2024] [Indexed: 04/19/2024] Open
Abstract
Renin-angiotensin system activation contributes to skeletal muscle atrophy in aging individuals with chronic diseases. We aimed to explore the effects of cholecalciferol (VD3) and calcitriol (1,25VD3) on signaling of muscle proteolysis and oxidative stress in myotubes challenged with angiotensin II (AII). The mouse C2C12 myotubes were assigned to vehicle, AII, AII + VD3, AII + 1,25VD3, and AII + losartan groups. The expression levels of muscle-specific E3 ubiquitin ligase proteins, autophagy-related proteins, and oxidative stress markers were investigated. We demonstrated the diverse effects of VD3 and 1,25VD3 on AII-induced myotube atrophy. The myotube diameter was preserved by treatment with 100 nM VD3 and losartan, while 1 and 10 nM 1,25VD3 increased levels of FoxO3a, MuRF1, and atrogin-1 protein expression in myotubes exposed to AII. Treatment with AII + 10 nM 1,25VD3 resulted in the upregulation of LC3B-II, LC3B-II/LC3B-I, and mature cathepsin L, which are autophagic marker proteins. The p62/SQSTM1 protein was downregulated and vitamin D receptor was upregulated after treatment with AII + 10 nM 1,25VD3. A cellular redox imbalance was observed as AII + 10 nM 1,25VD3-induced reactive oxygen species and NADPH oxidase-2 overproduction, and these changes were associated with an inadequate response of antioxidant superoxide dismutase-1 and catalase proteins. Collectively, these findings provide a translational perspective on the role of vitamin D3 in alleviating muscle atrophy related to high levels of AII.
Collapse
Affiliation(s)
- Muthita Hirunsai
- Department of Biopharmacy, Faculty of PharmacySrinakharinwirot UniversityNakhon NayokThailand
| | - Ratchakrit Srikuea
- Department of Physiology, Faculty of ScienceMahidol UniversityBangkokThailand
| |
Collapse
|
9
|
Chen W, Wang D, Ma L, Wu F, Ren Q, Tao J, Chen X, Zhang A. Chronic arsenite exposure induced skeletal muscle atrophy by disrupting angiotensin II-melatonin axis in rats. ENVIRONMENTAL TOXICOLOGY 2024; 39:1350-1359. [PMID: 37966059 DOI: 10.1002/tox.24027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/06/2023] [Accepted: 10/31/2023] [Indexed: 11/16/2023]
Abstract
Arsenic is a well-known environmental toxicant and emerging evidence suggests that arsenic exposure has potential skeletal muscle toxicity; however, the underlying mechanism has not yet been clarified. The aim of this study was to investigate the correlation among adverse effects of subchronic and chronic environmental arsenic exposure on skeletal muscle as well as specific myokines secretion and angiotensin II (AngII)-melatonin (MT) axis in rats. Four-week-old rats were exposed to arsenite (iAs) in drinking water at environmental relevant concentration of 10 ppm for 3 or 9 months. Results indicated that the gastrocnemius muscle had atrophied and its mass was decreased in rats exposed to arsenite for 9 months, whereas, they had no significant changes in rats exposed to arsenite for 3 months. The levels of serum-specific myokine irisin and gastrocnemius muscle insulin-like growth factor-1 (IGF-1) were increased in 3-month exposure group and decreased in 9-month exposure group, while serum myostatin (MSTN) was increased significantly in 9-month exposure group. In addition, serum AngII level increased both in 3- and 9-month exposure groups, while serum MT level increased in 3-month exposure group and decreased in 9-month exposure group. Importantly, the ratio of AngII to MT level in serum increased gradually with the prolongation of arsenite exposure. It showed a certain correlation between AngII-MT axis and gastrocnemius muscle mass, gastrocnemius muscle level of IGF-1 or serum levels of irisin and MSTN. In conclusion, the disruption of AngII-MT axis balance may be a significant factor for skeletal muscle atrophy induced by chronic environmental arsenic exposure.
Collapse
Affiliation(s)
- Wanying Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Department of Toxicology, Guizhou Medical University, Guiyang, China
| | - Dapeng Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Department of Toxicology, Guizhou Medical University, Guiyang, China
| | - Lu Ma
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Department of Toxicology, Guizhou Medical University, Guiyang, China
| | - Fan Wu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Department of Toxicology, Guizhou Medical University, Guiyang, China
| | - Qian Ren
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Department of Toxicology, Guizhou Medical University, Guiyang, China
| | - Junyan Tao
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Department of Toxicology, Guizhou Medical University, Guiyang, China
| | - Xiong Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Department of Toxicology, Guizhou Medical University, Guiyang, China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Department of Toxicology, Guizhou Medical University, Guiyang, China
| |
Collapse
|
10
|
Rossios C, Bashir T, Achison M, Adamson S, Akpan A, Aspray T, Avenell A, Band MM, Burton LA, Cvoro V, Donnan PT, Duncan GW, George J, Gordon AL, Gregson CL, Hapca A, Hume C, Jackson TA, Kerr S, Kilgour A, Masud T, McKenzie A, McKenzie E, Patel H, Pilvinyte K, Roberts HC, Sayer AA, Smith KT, Soiza RL, Steves CJ, Struthers AD, Tiwari D, Whitney J, Witham MD, Kemp PR. ACE I/D genotype associates with strength in sarcopenic men but not with response to ACE inhibitor therapy in older adults with sarcopenia: Results from the LACE trial. PLoS One 2023; 18:e0292402. [PMID: 37862321 PMCID: PMC10588903 DOI: 10.1371/journal.pone.0292402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/19/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Angiotensin II (AII), has been suggested to promote muscle loss. Reducing AII synthesis, by inhibiting angiotensin converting enzyme (ACE) activity has been proposed as a method to inhibit muscle loss. The LACE clinical trial was designed to determine whether ACE inhibition would reduce further muscle loss in individuals with sarcopenia but suffered from low recruitment and returned a negative result. Polymorphic variation in the ACE promoter (I/D alleles) has been associated with differences in ACE activity and muscle physiology in a range of clinical conditions. This aim of this analysis was to determine whether I/D polymorphic variation is associated with muscle mass, strength, in sarcopenia or contributed to the lack of response to treatment in the LACE study. METHODS Sarcopenic individuals were recruited into a 2x2 factorial multicentre double-blind study of the effects of perindopril and/or leucine versus placebo on physical performance and muscle mass. DNA extracted from blood samples (n = 130 72 women and 58 men) was genotyped by PCR for the ACE I/D polymorphism. Genotypes were then compared with body composition measured by DXA, hand grip and quadriceps strength before and after 12 months' treatment with leucine and/or perindopril in a cross-sectional analysis of the influence of genotype on these variables. RESULTS Allele frequencies for the normal UK population were extracted from 13 previous studies (I = 0.473, D = 0.527). In the LACE cohort the D allele was over-represented (I = 0.412, D = 0.588, p = 0.046). This over-representation was present in men (I = 0.353, D = 0.647, p = 0.010) but not women (I = 0.458, D = 0.532, p = 0.708). In men but not women, individuals with the I allele had greater leg strength (II/ID = 18.00 kg (14.50, 21.60) vs DD = 13.20 kg (10.50, 15.90), p = 0.028). Over the 12 months individuals with the DD genotype increased in quadriceps strength but those with the II or ID genotype did not. Perindopril did not increase muscle strength or mass in any polymorphism group relative to placebo. CONCLUSION Our results suggest that although ACE genotype was not associated with response to ACE inhibitor therapy in the LACE trial population, sarcopenic men with the ACE DD genotype may be weaker than those with the ACE I/D or II genotype.
Collapse
Affiliation(s)
- Christos Rossios
- Cardiovascular and Respiratory Interface Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Tufail Bashir
- Cardiovascular and Respiratory Interface Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Marcus Achison
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Simon Adamson
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Asangaedem Akpan
- University of Liverpool, Liverpool University Hospitals NHS FT Trust, Clinical Research Network Northwest Coast, Liverpool, United Kingdom
| | - Terry Aspray
- AGE Research Group, NIHR Newcastle Biomedical Research Centre, Translational Clinical Research Institute, Newcastle University, Cumbria Northumberland Tyne and Wear NHS Foundation Trust and Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Alison Avenell
- Health Services Research Unit, University of Aberdeen, Aberdeen, United Kingdom
| | - Margaret M. Band
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Louise A. Burton
- Medicine for the Elderly, NHS Tayside, Dundee, United Kingdom
- Ageing and Health, University of Dundee, Dundee, United Kingdom
| | - Vera Cvoro
- Victoria Hospital, Kirkcaldy, United Kingdom
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter T. Donnan
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Gordon W. Duncan
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Medicine for the Elderly, NHS Lothian, Edinburgh, United Kingdom
| | - Jacob George
- Dept Clinical Pharmacology, Division of Molecular & Clinical Medicine, University of Dundee Medical School, Ninewells Hospital, Dundee, United Kingdom
| | - Adam L. Gordon
- Unit of Injury, Inflammation and Recovery, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- NIHR Nottingham Biomedical Research Centre, Department of Medicine for the Elderly, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, United Kingdom
| | - Celia L. Gregson
- Musculoskeletal Research Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Older Person’s Unit, Royal United Hospital NHS Foundation Trust Bath, Bath, United Kingdom
| | - Adrian Hapca
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Cheryl Hume
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Thomas A. Jackson
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Simon Kerr
- Department of Older People’s Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Alixe Kilgour
- Medicine for the Elderly, NHS Lothian, Edinburgh, United Kingdom
- Ageing and Health Research Group, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Tahir Masud
- Clinical Gerontology Research Unit, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, United Kingdom
| | - Andrew McKenzie
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Emma McKenzie
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Harnish Patel
- NIHR Biomedical Research Centre, University of Southampton and University Hospital Southampton NHSFT, Southampton, Hampshire, United Kingdom
| | - Kristina Pilvinyte
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Helen C. Roberts
- Academic Geriatric Medicine, University of Southampton, Mailpoint 807 Southampton General Hospital, Southampton, United Kingdom
| | - Avan A. Sayer
- AGE Research Group, NIHR Newcastle Biomedical Research Centre, Translational Clinical Research Institute, Newcastle University, Cumbria Northumberland Tyne and Wear NHS Foundation Trust and Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Karen T. Smith
- Tayside Clinical Trials Unit (TCTU), Tayside Medical Science Centre (TASC), University of Dundee, Ninewells Hospital & Medical School, Dundee, United Kingdom
| | - Roy L. Soiza
- Ageing & Clinical Experimental Research (ACER) Group, University of Aberdeen, Aberdeen, United Kingdom
| | - Claire J. Steves
- Department of Twin Research and Genetic Epidemiology, King’s College London & Department of Clinical Gerontology, King’s College Hospital, London, United Kingdom
| | - Allan D. Struthers
- Dept Clinical Pharmacology, Division of Molecular & Clinical Medicine, University of Dundee Medical School, Ninewells Hospital, Dundee, United Kingdom
| | - Divya Tiwari
- Bournemouth University and Royal Bournemouth Hospital, Bournemouth, United Kingdom
| | - Julie Whitney
- School of Population Health & Environmental Sciences, King’s College London and King’s College Hospital, London, United Kingdom
| | - Miles D. Witham
- AGE Research Group, NIHR Newcastle Biomedical Research Centre, Translational Clinical Research Institute, Newcastle University, Cumbria Northumberland Tyne and Wear NHS Foundation Trust and Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Paul R. Kemp
- Cardiovascular and Respiratory Interface Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, United Kingdom
| |
Collapse
|
11
|
Coelho-Júnior HJ, de Oliveira Gonçalves I, Landi F, Calvani R, Tosato M, Picca A, Marzetti E. Muscle power-related parameters in middle-aged and older Brazilian women: a cross-sectional study. Sci Rep 2023; 13:13186. [PMID: 37580323 PMCID: PMC10425341 DOI: 10.1038/s41598-023-39182-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/20/2023] [Indexed: 08/16/2023] Open
Abstract
The present study was conducted to provide normative values for lower-limb muscle power estimated through equations based on the 5 times sit-to-stand (5STS) test in Brazilian older women. In addition, we investigated the association between muscle power parameters and age. The study followed a cross-sectional design. Participants were community-dwelling women. Candidates were considered eligible if they were 18 years or older, lived independently, and possessed sufficient physical and cognitive abilities to perform all measurements required by the protocol. The 5STS test was performed as fast as possible using a standard protocol. Absolute, relative, and allometric muscle power measures were estimated using 5STS-based equations. Two thousand four-hundred seventy-one women participated in the present study. Results indicated that muscle power-related parameters decreased linearly with age. Women 60-69 years showed a marginal reduction in absolute (- 5.2%), relative (- 7.9%), and allometric (- 4.0%) muscle power. A larger reduction was observed in those 70-79 years and reached ¼ of loss in participants ≥ 80, in comparison to middle-aged participants. Pearson's correlation and linear regression analyses indicated that power-related parameters were negatively associated with age. In conclusion, data of the present study provide normative values for lower-limb muscle power parameters according to 5STS-based equations. We observed that muscle power-related parameters declined with age, such that participants 60-69, 70-79, and ≥ 80 years displayed lower absolute and relative muscle power compared middle-aged women. A later decline was observed in allometric muscle power. Relative muscle power declined to a greater extent than other parameters, suggesting a possible window of opportunity for interventions.
Collapse
Affiliation(s)
- Hélio José Coelho-Júnior
- Department of Geriatrics, Orthopedics and Rheumatology, Center for Geriatric Medicine (Ce.M.I.), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | | | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Center for Geriatric Medicine (Ce.M.I.), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy
| | - Matteo Tosato
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy
- Department of Medicine and Surgery, LUM University, SS100 km 18, 70010, Casamassima, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Center for Geriatric Medicine (Ce.M.I.), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy.
| |
Collapse
|
12
|
Wang T, Xu H, Wu S, Guo Y, Zhao G, Wang D. Mechanisms Underlying the Effects of the Green Tea Polyphenol EGCG in Sarcopenia Prevention and Management. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37316469 DOI: 10.1021/acs.jafc.3c02023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Sarcopenia is prevalent among the older population and severely affects human health. Tea catechins may benefit for skeletal muscle performance and protect against secondary sarcopenia. However, the mechanisms underlying their antisarcopenic effect are still not fully understood. Despite initial successes in animal and early clinical trials regarding the safety and efficacy of (-)-epigallocatechin-3-gallate (EGCG), a major catechin of green tea, many challenges, problems, and unanswered questions remain. In this comprehensive review, we discuss the potential role and underlying mechanisms of EGCG in sarcopenia prevention and management. We thoroughly review the general biological activities and general effects of EGCG on skeletal muscle performance, EGCG's antisarcopenic mechanisms, and recent clinical evidence of the aforesaid effects and mechanisms. We also address safety issues and provide directions for future studies. The possible concerted actions of EGCG indicate the need for further studies on sarcopenia prevention and management in humans.
Collapse
Affiliation(s)
- Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, 212000 Zhenjiang, China
| | - Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China
| | - Shanshan Wu
- College of Agriculture & Biotechnology, Zhejiang University, 310058 Hangzhou, China
| | - Yuanxin Guo
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China
| | - Guangshan Zhao
- College of Food Science & Technology, Henan Agricultural University, 450002 Zhengzhou, China
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, 212100 Zhenjiang, China
| |
Collapse
|
13
|
Habibian JS, Bolino MJ, Ferguson BS. HDAC8 regulates protein kinase D phosphorylation in skeletal myoblasts in response to stress signaling. Biochem Biophys Res Commun 2023; 650:81-86. [PMID: 36773343 PMCID: PMC9975084 DOI: 10.1016/j.bbrc.2023.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/15/2022] [Accepted: 02/02/2023] [Indexed: 02/08/2023]
Abstract
Skeletal muscle differentiation involves activation of quiescent satellite cells to proliferate, differentiate and fuse to form new myofibers; this requires coordination of myogenic transcription factors. Myogenic transcription is tightly regulated by various intracellular signaling pathways, which include members of the protein kinase D (PKD) family. PKD is a family of serine-threonine kinases that regulate gene expression, protein secretion, cell proliferation, differentiation and inflammation. PKD is a unique PKC family member that shares distant sequence homology to calcium-regulated kinases and plays an important role in muscle physiology. In this report, we show that class I histone deacetylase (HDAC) inhibition, and in particular HDAC8 inhibition, attenuated PKD phosphorylation in skeletal C2C12 myoblasts in response to phorbol ester, angiotensin II and dexamethasone signaling independent of changes in total PKD protein expression. As class I HDACs and PKD signaling are requisite for myocyte differentiation, these data suggest that HDAC8 functions as a potential feedback regulator of PKD phosphorylation to control myogenic gene expression.
Collapse
Affiliation(s)
- Justine S Habibian
- Department of Nutrition, The University of Nevada Reno, Reno, NV, 89557, USA; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV, 89557, USA
| | - Matthew J Bolino
- Department of Nutrition, The University of Nevada Reno, Reno, NV, 89557, USA; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV, 89557, USA
| | - Bradley S Ferguson
- Department of Nutrition, The University of Nevada Reno, Reno, NV, 89557, USA; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV, 89557, USA; Center of Biomedical Research Excellence for Molecular and Cellular Signal Transduction in the Cardiovascular System, The University of Nevada Reno, Reno, NV, 89557, USA.
| |
Collapse
|
14
|
Is the anti-aging effect of ACE2 due to its role in the renin-angiotensin system?-Findings from a comparison of the aging phenotypes of ACE2-deficient, Tsukuba hypertensive, and Mas-deficient mice. Hypertens Res 2023; 46:1210-1220. [PMID: 36788301 PMCID: PMC9925940 DOI: 10.1038/s41440-023-01189-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 02/16/2023]
Abstract
Angiotensin converting enzyme 2 (ACE2) functions as an enzyme that produces angiotensin 1-7 (A1-7) from angiotensin II (AII) in the renin-angiotensin system (RAS). We evaluated aging phenotypes, especially skeletal muscle aging, in ACE2 systemically deficient (ACE2 KO) mice and found that ACE2 has an antiaging function. The characteristic aging phenotype observed in ACE2 KO mice was not reproduced in mice deficient in the A1-7 receptor Mas or in Tsukuba hypertensive mice, a model of chronic AII overproduction, suggesting that ACE2 has a RAS-independent antiaging function. In this review, the results we have obtained and related studies on the aging regulatory mechanism mediated by RAS components will be presented and summarized. We evaluated the aging phenotype of ACE2 systemically deficient (ACE2 KO) mice, particularly skeletal muscle aging, and found that ACE2 has an antiaging function. The characteristic aging phenotype observed in ACE2 KO mice was not reproduced in Mas KO mice, angiotensin 1-7 receptor-deficient mice or in Tsukuba hypertensive mice, a model of chronic angiotensin II overproduction, suggesting that the antiaging functions of ACE2 are independent of the renin-angiotensin system (RAS).
Collapse
|
15
|
Holder ER, Alibhai FJ, Caudle SL, McDermott JC, Tobin SW. The importance of biological sex in cardiac cachexia. Am J Physiol Heart Circ Physiol 2022; 323:H609-H627. [PMID: 35960634 DOI: 10.1152/ajpheart.00187.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac cachexia is a catabolic muscle wasting syndrome observed in approximately 1 in 10 heart failure patients. Increased skeletal muscle atrophy leads to frailty and limits mobility which impacts quality of life, exacerbates clinical care, and is associated with higher rates of mortality. Heart failure is known to exhibit a wide range of prevalence and severity when examined across individuals of different ages and with co-morbidities related to diabetes, renal failure and pulmonary dysfunction. It is also recognized that men and women exhibit striking differences in the pathophysiology of heart failure as well as skeletal muscle homeostasis. Given that both skeletal muscle and heart failure physiology are in-part sex dependent, the diagnosis and treatment of cachexia in heart failure patients may depend on a comprehensive examination of how these organs interact. In this review we explore the potential for sex-specific differences in cardiac cachexia. We summarize advantages and disadvantages of clinical methods used to measure muscle mass and function and provide alternative measurements that should be considered in preclinical studies. Additionally, we summarize sex-dependent effects on muscle wasting in preclinical models of heart failure, disuse, and cancer. Lastly, we discuss the endocrine function of the heart and outline unanswered questions that could directly impact patient care.
Collapse
|
16
|
Sanz B, Rezola-Pardo C, Arrieta H, Fernández-Atutxa A, Lora-Diaz I, Gil-Goikouria J, Rodriguez-Larrad A, Irazusta J. High serum angiotensin-converting enzyme 2 activity as a biomarker of frailty in nursing home residents. Exp Gerontol 2021; 158:111655. [PMID: 34915109 DOI: 10.1016/j.exger.2021.111655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/19/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023]
Abstract
Angiotensin-converting enzyme (ACE) and angiotensin-converting enzyme 2 (ACE2) are two of the main components of the renin-angiotensin system (RAS). Imbalanced RAS showing lower ACE2 has been associated with increased cardiovascular risk, muscular pathologies, sarcopenia, frailty, other age-related pathologies and a poorer health status. However, its role in aging remains unclear. Thus, the aim of this work was to analyze the serum enzymatic activity of ACE and ACE2, the ACE/ACE2 ratio and its association with anthropometric parameters, blood pressure, physical function, dependence and frailty in older people living in nursing homes. This study is a secondary analysis of baseline data from two randomized clinical trials in a population of 228 older individuals living in nursing homes (Spain). Serum ACE and ACE2 enzymatic activities were measured by fluorimetry. Variables linked to cardiovascular risk, physical function, dependence and frailty were measured using validated tests, indexes and scales. Association between ACE, ACE2 serum activities, the ACE/ACE2 ratio and the rest of the quantitative variables were assessed by Pearson's correlations and by partial correlations controlled by age and sex. The association between serum ACE and ACE2 activities, the ACE/ACE2 ratio and frailty scores was analyzed by generalized linear models with and without controlling for sex and age. Differences in enzymatic activities between sexes and between frail and non-frail individuals were analyzed using Student's t-test and general linear models to control analysis by age and sex. We found that higher serum ACE2 activity was associated with a higher body mass index, worse physical function, greater dependence and increased frailty. This association is consistent with the elevation of circulating ACE2 in certain pathological conditions and in line with RAS deregulation in muscular dystrophies. Serum ACE2 activity, in combination with other molecules, could be proposed as a biomarker of poor physical function, higher dependence and frailty.
Collapse
Affiliation(s)
- Begoña Sanz
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain; Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain.
| | - Chloe Rezola-Pardo
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain.
| | - Haritz Arrieta
- Department of Nursing II, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 20014 Donostia-San Sebastián, Gipuzkoa, Spain.
| | - Ainhoa Fernández-Atutxa
- Department of Nursing I, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain.
| | - Inmaculada Lora-Diaz
- DomusVi Berra, Berratxo Bidea, 2, 20017 Donostia-San Sebastián, Gipuzkoa, Spain.
| | - Javier Gil-Goikouria
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain; Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain.
| | - Ana Rodriguez-Larrad
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain; Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain.
| | - Jon Irazusta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain; Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain.
| |
Collapse
|
17
|
Joshi B, Wagh G, Kaur H, Patra C. Zebrafish Model to Study Angiotensin II-Mediated Pathophysiology. BIOLOGY 2021; 10:1177. [PMID: 34827169 PMCID: PMC8614710 DOI: 10.3390/biology10111177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/31/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022]
Abstract
Hypertension, a common chronic condition, may damage multiple organs, including the kidney, heart, and brain. Thus, it is essential to understand the pathology upon ectopic activation of the molecular pathways involved in mammalian hypertension to develop strategies to manage hypertension. Animal models play a crucial role in unraveling the disease pathophysiology by allowing incisive experimental procedures impossible in humans. Zebrafish, a small freshwater fish, have emerged as an important model system to study human diseases. The primary effector, Angiotensin II of the RAS pathway, regulates hemodynamic pressure overload mediated cardiovascular pathogenesis in mammals. There are various established mammalian models available to study pathophysiology in Angiotensin II-induced hypertension. Here, we have developed a zebrafish model to study pathogenesis by Angiotensin II. We find that intradermal Angiotensin II injection every 12 h can induce cardiac remodeling in seven days. We show that Angiotensin II injection in adult zebrafish causes cardiomyocyte hypertrophy and enhances cardiac cell proliferation. In addition, Angiotensin II induces ECM protein-coding gene expression and fibrosis in the cardiac ventricles. Thus, this study can conclude that Angiotensin II injection in zebrafish has similar implications as mammals, and zebrafish can be a model to study pathophysiology associated with AngII-RAS signaling.
Collapse
Affiliation(s)
- Bhagyashri Joshi
- Developmental Biology, Agharkar Research Institute, Pune 411004, India; (B.J.); (G.W.)
- Science and Technology, SP Pune University, Pune 411007, India
| | - Ganesh Wagh
- Developmental Biology, Agharkar Research Institute, Pune 411004, India; (B.J.); (G.W.)
- Science and Technology, SP Pune University, Pune 411007, India
| | - Harmandeep Kaur
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5C 2T2, Canada;
| | - Chinmoy Patra
- Developmental Biology, Agharkar Research Institute, Pune 411004, India; (B.J.); (G.W.)
- Science and Technology, SP Pune University, Pune 411007, India
| |
Collapse
|
18
|
Angiotensin II inhibition: a potential treatment to slow the progression of sarcopenia. Clin Sci (Lond) 2021; 135:2503-2520. [PMID: 34751393 DOI: 10.1042/cs20210719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/21/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023]
Abstract
Sarcopenia is defined as the progressive and generalized loss of skeletal muscle mass and strength, which is associated with increased likelihood of adverse outcomes including falls, fractures, physical disability, and mortality. The etiology of sarcopenia has been postulated to be multifactorial with genetics, aging, immobility, nutritional deficiencies, inflammation, stress, and endocrine factors all contributing to the imbalance of muscle anabolism and catabolism. The prevalence of sarcopenia is estimated to range from 13 to 24% in adults over 60 years of age and up to 50% in persons aged 80 and older. As the population continues to age, the prevalence of sarcopenia continues to increase and is expected to affect 500 million people by the year 2050. Sarcopenia impacts the overall health of patients through limitations in functional status, increase in hospital readmissions, poorer hospital outcomes, and increase in overall mortality. Thus, there exists a need to prevent or reduce the occurrence of sarcopenia. Here, we explore the potential mechanisms and current studies regarding angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme (ACE) inhibitors on reducing the development of sarcopenia through the associated changes in cardiovascular function, renal function, muscle fiber composition, inflammation, endothelial dysfunction, metabolic efficiency, and mitochondrial function.
Collapse
|
19
|
Wilburn D, Ismaeel A, Machek S, Fletcher E, Koutakis P. Shared and distinct mechanisms of skeletal muscle atrophy: A narrative review. Ageing Res Rev 2021; 71:101463. [PMID: 34534682 DOI: 10.1016/j.arr.2021.101463] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 12/15/2022]
Abstract
Maintenance of skeletal muscle mass and function is an incredibly nuanced balance of anabolism and catabolism that can become distorted within different pathological conditions. In this paper we intend to discuss the distinct intracellular signaling events that regulate muscle protein atrophy for a given clinical occurrence. Aside from the common outcome of muscle deterioration, several conditions have at least one or more distinct mechanisms that creates unique intracellular environments that facilitate muscle loss. The subtle individuality to each of these given pathologies can provide both researchers and clinicians with specific targets of interest to further identify and increase the efficacy of medical treatments and interventions.
Collapse
Affiliation(s)
- Dylan Wilburn
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Ahmed Ismaeel
- Department of Biology, Baylor University, Waco, TX 76706, USA
| | - Steven Machek
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Emma Fletcher
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; Department of Biology, Baylor University, Waco, TX 76706, USA
| | | |
Collapse
|
20
|
Nguyen BL, Yoshihara T, Deminice R, Lawrence J, Ozdemir M, Hyatt H, Powers SK. Alterations in renin-angiotensin receptors are not responsible for exercise preconditioning of skeletal muscle fibers. SPORTS MEDICINE AND HEALTH SCIENCE 2021; 3:148-156. [PMID: 35784524 PMCID: PMC9219300 DOI: 10.1016/j.smhs.2021.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 11/27/2022] Open
Abstract
Endurance exercise training promotes a protective phenotype in skeletal muscle known as exercise preconditioning. Exercise preconditioning protects muscle fibers against a variety of threats including inactivity-induced muscle atrophy. The mechanism(s) responsible for exercise preconditioning remain unknown and are explored in these experiments. Specifically, we investigated the impact of endurance exercise training on key components of the renin-angiotensin system (RAS). The RAS was targeted because activation of the classical axis of the RAS pathway via angiotensin II type I receptors (AT1Rs) promotes muscle atrophy whereas activation of the non-classical RAS axis via Mas receptors (MasRs) inhibits the atrophic signaling of the classical RAS pathway. Guided by prior studies, we hypothesized that an exercise-induced decrease in AT1Rs and/or increases in MasRs in skeletal muscle fibers is a potential mechanism responsible for exercise preconditioning. Following endurance exercise training in rats, we examined the abundance of AT1Rs and MasRs in both locomotor and respiratory muscles. Our results indicate that endurance exercise training does not alter the protein abundance of AT1Rs or MasRs in muscle fibers from the diaphragm, plantaris, and soleus muscles compared to sedentary controls (p > 0.05). Furthermore, fluorescent angiotensin II (AngII) binding analyses confirm our results that exercise preconditioning does not alter the protein abundance of AT1Rs in the diaphragm, plantaris, and soleus (p > 0.05). This study confirms that exercise-induced changes in RAS receptors are not a key mechanism that contributes to the beneficial effects of exercise preconditioning in skeletal muscle fibers.
Collapse
Affiliation(s)
- Branden L. Nguyen
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, 32611, Florida, USA
| | - Toshinori Yoshihara
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, 32611, Florida, USA
- Department of Exercise Physiology, Juntendo University, Tokyo, 270-1695, Japan
| | - Rafael Deminice
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, 32611, Florida, USA
- Department of Physical Education, University of Estadual of Londrina, Londrina, 10011, Brazil
| | - Jensen Lawrence
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, 32611, Florida, USA
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, 32611, Florida, USA
| | - Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, 32611, Florida, USA
| | - Scott K. Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, 32611, Florida, USA
- Corresponding authors. Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Heart failure (HF) is a structural or functional cardiac abnormality which leads to failure of the heart to deliver oxygen commensurately with the requirements of the tissues and it may progress to a generalized wasting of skeletal muscle, fat tissue, and bone tissue (cardiac cachexia). Clinically, dyspnea, fatigue, and exercise intolerance are some typical signs and symptoms that characterize HF patients. This review focused on the phenotypic characteristics of HF-induced skeletal myopathy as well as the mechanisms of muscle wasting due to HF and highlighted possible therapeutic strategies for skeletal muscle wasting in HF. RECENT FINDINGS The impaired exercise capacity of those patients is not attributed to the reduced blood flow in the exercising muscles, but rather to abnormal metabolic responses, myocyte apoptosis and atrophy of skeletal muscle. Specifically, the development of skeletal muscle wasting in chronic HF is characterized by structural, metabolic, and functional abnormalities in skeletal muscle and may be a result not only of reduced physical activity, but also of metabolic or hormonal derangements that favour catabolism over anabolism. In particular, abnormal energy metabolism, mitochondrial dysfunction, transition of myofibers from type I to type II, muscle atrophy, and reduction in muscular strength are included in skeletal muscle abnormalities which play a central role in the decreased exercise capacity of HF patients. Skeletal muscle alterations and exercise intolerance observed in HF are reversible by exercise training, since it is the only demonstrated intervention able to improve skeletal muscle metabolism, growth factor activity, and functional capacity and to reverse peripheral abnormalities.
Collapse
|
22
|
Adams V, Wunderlich S, Mangner N, Hommel J, Esefeld K, Gielen S, Halle M, Ellingsen Ø, Van Craenenbroeck EM, Wisløff U, Pieske B, Linke A, Winzer EB. Ubiquitin-proteasome-system and enzymes of energy metabolism in skeletal muscle of patients with HFpEF and HFrEF. ESC Heart Fail 2021; 8:2556-2568. [PMID: 33955206 PMCID: PMC8318515 DOI: 10.1002/ehf2.13405] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/17/2021] [Accepted: 04/22/2021] [Indexed: 01/14/2023] Open
Abstract
Background Skeletal muscle (SM) alterations contribute to exercise intolerance in heart failure patients with preserved (HFpEF) or reduced (HFrEF) left ventricular ejection fraction (LVEF). Protein degradation via the ubiquitin‐proteasome‐system (UPS), nuclear apoptosis, and reduced mitochondrial energy supply is associated with SM weakness in HFrEF. These mechanisms are incompletely studied in HFpEF, and a direct comparison between these groups is missing. Methods and results Patients with HFpEF (LVEF ≥ 50%, septal E/e′ > 15 or >8 and NT‐proBNP > 220 pg/mL, n = 20), HFrEF (LVEF ≤ 35%, n = 20) and sedentary control subjects (Con, n = 12) were studied. Inflammatory markers were measured in serum, and markers of the UPS, nuclear apoptosis, and energy metabolism were determined in percutaneous SM biopsies. Both HFpEF and HFrEF showed increased proteolysis (MuRF‐1 protein expression, ubiquitination, and proteasome activity) with proteasome activity significantly related to interleukin‐6. Proteolysis was more pronounced in patients with lower exercise capacity as indicated by peak oxygen uptake in per cent predicted below the median. Markers of apoptosis did not differ between groups. Mitochondrial energy supply was reduced in HFpEF and HFrEF (complex‐I activity: −31% and −53%; malate dehydrogenase activity: −20% and −29%; both P < 0.05 vs. Con). In contrast, short‐term energy supply via creatine kinase was increased in HFpEF but decreased in HFrEF (47% and −45%; P < 0.05 vs. Con). Conclusions Similarly to HFrEF, skeletal muscle in HFpEF is characterized by increased proteolysis linked to systemic inflammation and reduced exercise capacity. Energy metabolism is disturbed in both groups; however, its regulation seems to be severity‐dependent.
Collapse
Affiliation(s)
- Volker Adams
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany.,Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| | - Sebastian Wunderlich
- Department of Internal Medicine/Cardiology, Heart Center Leipzig - University Hospital, Leipzig, Germany
| | - Norman Mangner
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| | - Jennifer Hommel
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| | - Katrin Esefeld
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Prevention and Sports Medicine, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Stephan Gielen
- Department of Cardiology, Angiology and Intensive Care, Klinikum Lippe, Detmold, Germany
| | - Martin Halle
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Prevention and Sports Medicine, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Øyvind Ellingsen
- Department of Cardiology, St. Olavs University Hospital, Trondheim, Norway.,The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Emeline M Van Craenenbroeck
- Department of Cardiology, Antwerp University Hospital, Edegem, Belgium.,Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Ulrik Wisløff
- The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Burkert Pieske
- Department Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany.,Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| | - Ephraim B Winzer
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Heart Center Dresden - University Hospital, Herzzentrum Dresden, Universitätsklinik, Fetscherstraße 76, Dresden, 01307, Germany
| |
Collapse
|
23
|
Frailty is not associated with hypertension, blood pressure or antihypertensive medication in community-dwelling older adults: A cross-sectional comparison across 3 frailty instruments. Exp Gerontol 2021; 146:111245. [PMID: 33476700 DOI: 10.1016/j.exger.2021.111245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/04/2021] [Accepted: 01/13/2021] [Indexed: 12/29/2022]
Abstract
AIM The present study investigated whether hypertension, blood pressure, and antihypertensive therapy were associated with frailty status in community-dwelling older adults. In addition, we tested whether such associations were consistent across different frailty instruments. MATERIAL AND METHODS Two-hundred older adults were enrolled in the study. Participant frailty status was determined according to a modified physical frailty phenotype (mFP), the FRAIL scale, and the Study of Osteoporotic Fracture (SOF) index. Blood pressure was assessed three times, in three different days, and mean values were used in the final analysis. Information pertaining to disease conditions and antihypertensive therapy were collected by two researchers through self-report and careful review of medical charts. RESULTS No significant differences in hemodynamic parameters, hypertension diagnosis, and antihypertensive therapy were observed across frailty statuses, regardless of the frailty assessment tool used. CONCLUSION Findings of the present study indicate that hypertension, blood pressure levels and antihypertensive medication were not cross-sectionally associated with frailty status in cognitively preserved community-dwelling older adults with low prevalence of comorbidities, regardless of the tool used for frailty identification.
Collapse
|
24
|
Pinto PR, Yoshinaga MY, Del Bianco V, Bochi AP, Ferreira GS, Pinto IFD, Rodrigues LG, Nakandakare ER, Okamoto MM, Machado UF, Miyamoto S, Catanozi S, Passarelli M. Dietary sodium restriction alters muscle lipidomics that relates to insulin resistance in mice. J Biol Chem 2021; 296:100344. [PMID: 33524391 PMCID: PMC7949138 DOI: 10.1016/j.jbc.2021.100344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
A low-sodium (LS) diet has been shown to reduce blood pressure (BP) and the incidence of cardiovascular diseases. However, severe dietary sodium restriction promotes insulin resistance (IR) and dyslipidemia in animal models and humans. Thus, further clarification of the long-term consequences of LS is needed. Here, we investigated the effects of chronic LS on gastrocnemius gene and protein expression and lipidomics and its association with IR and plasma lipids in LDL receptor knockout mice. Three-month-old male mice were fed a normal sodium diet (NS; 0.5% Na; n = 12-19) or LS (0.06% Na; n = 14-20) over 90 days. Body mass (BM), BP, plasma total cholesterol, triacylglycerol (TG), glucose, hematocrit, and IR were evaluated. LS increased BM (9%), plasma TG (51%), blood glucose (19%), and IR (46%) when compared with the NS. RT-qPCR analysis revealed that genes involved in lipid uptake and oxidation were increased by the LS: Fabp3 (106%), Prkaa1 (46%), and Cpt1 (74%). Genes and proteins (assessed by Western blotting) involved in insulin signaling were not changed by the LS. Similarly, lipid species classically involved in muscle IR, such as diacylglycerols and ceramides detected by ultra-high-performance liquid chromatography coupled to mass spectrometry, were also unchanged by LS. Species of phosphatidylcholines (68%), phosphatidylinositol (90%), and free fatty acids (59%) increased while cardiolipins (41%) and acylcarnitines (9%) decreased in gastrocnemius in response to LS and were associated with glucose disposal rate. Together these results suggest that chronic LS alters glycerophospholipid and fatty acids species in gastrocnemius that may contribute to glucose and lipid homeostasis derangements in mice.
Collapse
Affiliation(s)
- Paula Ramos Pinto
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marcos Y Yoshinaga
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Vanessa Del Bianco
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Paula Bochi
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Guilherme S Ferreira
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Isabella F D Pinto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Letícia G Rodrigues
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Edna R Nakandakare
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maristela M Okamoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ubiratan F Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Catanozi
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marisa Passarelli
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil.
| |
Collapse
|
25
|
Armstrong VS, Fitzgerald LW, Bathe OF. Cancer-Associated Muscle Wasting-Candidate Mechanisms and Molecular Pathways. Int J Mol Sci 2020; 21:ijms21239268. [PMID: 33291708 PMCID: PMC7729509 DOI: 10.3390/ijms21239268] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Excessive muscle loss is commonly observed in cancer patients and its association with poor prognosis has been well-established. Cancer-associated sarcopenia differs from age-related wasting in that it is not responsive to nutritional intervention and exercise. This is related to its unique pathogenesis, a result of diverse and interconnected mechanisms including inflammation, disordered metabolism, proteolysis and autophagy. There is a growing body of evidence that suggests that the tumor is the driver of muscle wasting by its elaboration of mediators that influence each of these pro-sarcopenic pathways. In this review, evidence for these tumor-derived factors and putative mechanisms for inducing muscle wasting will be reviewed. Potential targets for future research and therapeutic interventions will also be reviewed.
Collapse
Affiliation(s)
- Victoria S. Armstrong
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Liam W. Fitzgerald
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Oliver F. Bathe
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Departments of Surgery and Oncology, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Correspondence: ; Tel.: +1-403-521-3275
| |
Collapse
|
26
|
Webster JM, Kempen LJAP, Hardy RS, Langen RCJ. Inflammation and Skeletal Muscle Wasting During Cachexia. Front Physiol 2020; 11:597675. [PMID: 33329046 PMCID: PMC7710765 DOI: 10.3389/fphys.2020.597675] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
Cachexia is the involuntary loss of muscle and adipose tissue that strongly affects mortality and treatment efficacy in patients with cancer or chronic inflammatory disease. Currently, no specific treatments or interventions are available for patients developing this disorder. Given the well-documented involvement of pro-inflammatory cytokines in muscle and fat metabolism in physiological responses and in the pathophysiology of chronic inflammatory disease and cancer, considerable interest has revolved around their role in mediating cachexia. This has been supported by association studies that report increased levels of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) in some, but not all, cancers and in chronic inflammatory diseases such as chronic obstructive pulmonary disease (COPD) and rheumatoid arthritis (RA). In addition, preclinical studies including animal disease models have provided a substantial body of evidence implicating a causal contribution of systemic inflammation to cachexia. The presence of inflammatory cytokines can affect skeletal muscle through several direct mechanisms, relying on activation of the corresponding receptor expressed by muscle, and resulting in inhibition of muscle protein synthesis (MPS), elevation of catabolic activity through the ubiquitin-proteasomal system (UPS) and autophagy, and impairment of myogenesis. Additionally, systemic inflammatory mediators indirectly contribute to muscle wasting through dysregulation of tissue and organ systems, including GCs via the hypothalamus-pituitary-adrenal (HPA) axis, the digestive system leading to anorexia-cachexia, and alterations in liver and adipocyte behavior, which subsequently impact on muscle. Finally, myokines secreted by skeletal muscle itself in response to inflammation have been implicated as autocrine and endocrine mediators of cachexia, as well as potential modulators of this debilitating condition. While inflammation has been shown to play a pivotal role in cachexia development, further understanding how these cytokines contribute to disease progression is required to reveal biomarkers or diagnostic tools to help identify at risk patients, or enable the design of targeted therapies to prevent or delay the progression of cachexia.
Collapse
Affiliation(s)
- Justine M. Webster
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Laura J. A. P. Kempen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Rowan S. Hardy
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Institute for Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Ramon C. J. Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
27
|
Gupta S, Mitra A. Heal the heart through gut (hormone) ghrelin: a potential player to combat heart failure. Heart Fail Rev 2020; 26:417-435. [PMID: 33025414 DOI: 10.1007/s10741-020-10032-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Ghrelin, a small peptide hormone (28 aa), secreted mainly by X/A-like cells of gastric mucosa, is also locally produced in cardiomyocytes. Being an orexigenic factor (appetite stimulant), it promotes release of growth hormone (GH) and exerts diverse physiological functions, viz. regulation of energy balance, glucose, and/or fat metabolism for body weight maintenance. Interestingly, administration of exogenous ghrelin significantly improves cardiac functions in CVD patients as well as experimental animal models of heart failure. Ghrelin ameliorates pathophysiological condition of the heart in myocardial infarction, cardiac hypertrophy, fibrosis, cachexia, and ischemia reperfusion injury. This peptide also exerts significant impact at the level of vasculature leading to lowering high blood pressure and reversal of endothelial dysfunction and atherosclerosis. However, the molecular mechanism of actions elucidating the healing effects of ghrelin on the cardiovascular system is still a matter of conjecture. Some experimental data indicate its beneficial effects via complex cellular cross talks between autonomic nervous system and cardiovascular cells, some other suggest more direct receptor-mediated molecular actions via autophagy or ionotropic regulation and interfering with apoptotic and inflammatory pathways of cardiomyocytes and vascular endothelial cells. Here, in this review, we summarise available recent data to encourage more research to find the missing links of unknown ghrelin receptor-mediated pathways as we see ghrelin as a future novel therapy in cardiovascular protection.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, Paschim Bardhaman, 713347, India
| | - Arkadeep Mitra
- Department of Zoology, City College , 102/1, Raja Rammohan Sarani, Kolkata, 700009, India.
| |
Collapse
|
28
|
Aguirre F, Abrigo J, Gonzalez F, Gonzalez A, Simon F, Cabello-Verrugio C. Protective Effect of Angiotensin 1-7 on Sarcopenia Induced by Chronic Liver Disease in Mice. Int J Mol Sci 2020; 21:ijms21113891. [PMID: 32485991 PMCID: PMC7312494 DOI: 10.3390/ijms21113891] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/23/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia associated with chronic liver disease (CLD) is one of the more common extrahepatic features in patients with these pathologies. Among the cellular alterations observed in the muscle tissue under CLD is the decline in the muscle strength and function, as well as the increased fatigue. Morphological changes, such as a decrease in the fiber diameter and transition in the fiber type, are also reported. At the molecular level, sarcopenia for CLD is characterized by: (i) a decrease in the sarcomeric protein, such as myosin heavy chain (MHC); (ii) an increase in the ubiquitin–proteasome system markers, such as atrogin-1/MAFbx1 and MuRF-1/TRIM63; (iii) an increase in autophagy markers, such as LC3II/LC3I ratio. Among the regulators of muscle mass is the renin-angiotensin system (RAS). The non-classical axis of RAS includes the Angiotensin 1–7 [Ang-(1-7)] peptide and its receptor Mas, which in skeletal muscle has anti-atrophic effect in models of muscle wasting induced by immobilization, lipopolysaccharide, myostatin or angiotensin II. In this paper, we evaluated the effect of Ang-(1-7) on the sarcopenia by CLD in a murine model induced by the 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) hepatotoxin administered through diet. Our results show that Ang-(1-7) administration prevented the decline of the function and strength of muscle and increased the fatigue detected in the DDC-fed mice. Besides, we observed that the decreased fiber diameter and MHC levels, as well as the transition of fiber types, were all abolished by Ang-(1-7) in mice fed with DDC. Finally, Ang-(1-7) can decrease the atrogin-1 and MuRF-1 expression as well as the autophagy marker in mice treated with DDC. Together, our data support the protective role of Ang-(1-7) on the sarcopenia by CLD in mice.
Collapse
Affiliation(s)
- Francisco Aguirre
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile; (F.A.); (J.A.); (F.G.); (A.G.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile;
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Johanna Abrigo
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile; (F.A.); (J.A.); (F.G.); (A.G.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile;
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Francisco Gonzalez
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile; (F.A.); (J.A.); (F.G.); (A.G.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile;
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Andrea Gonzalez
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile; (F.A.); (J.A.); (F.G.); (A.G.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile;
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile;
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8370146, Chile
- Laboratory of Integrative Physiopathology, Department of Biological Science, Faculty of Life Science, Universidad Andres Bello, Santiago 8370146, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile; (F.A.); (J.A.); (F.G.); (A.G.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile;
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
- Correspondence: ; Tel./Fax: +56227703665
| |
Collapse
|
29
|
Liu Y, Bi X, Zhang Y, Wang Y, Ding W. Mitochondrial dysfunction/NLRP3 inflammasome axis contributes to angiotensin II-induced skeletal muscle wasting via PPAR-γ. J Transl Med 2020; 100:712-726. [PMID: 31857693 DOI: 10.1038/s41374-019-0355-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 10/19/2019] [Accepted: 11/04/2019] [Indexed: 12/15/2022] Open
Abstract
Angiotensin II (Ang II) levels are elevated in patients with chronic kidney disease or heart failure, and directly causes skeletal muscle wasting in rodents, but the molecular mechanisms of Ang II-induced skeletal muscle wasting and its potential as a therapeutic target are unknown. We investigated the NLR family pyrin domain containing 3 (NLRP3) inflammasome-mediated muscle atrophy response to Ang II in C2C12 myotubes and Nlrp3 knockout mice. We also assessed the mitochondrial dysfunction (MtD)/NLRP3 inflammasome axis in Ang II-induced C2C12 myotubes. Finally, we examined whether a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist could attenuate skeletal muscle wasting by targeting the MtD/NLRP3 inflammasome axis in vitro and in vivo. We demonstrated that Ang II increased NLRP3 inflammasome activation in cultured C2C12 myotubes dose dependently. Nlrp3 knockdown or Nlrp3-/- mice were protected from the imbalance of protein synthesis and degradation. Exposure of C2C12 to Ang II increased mitochondrial ROS (mtROS) generation, accompanied by MtD. Remarkably, the mitochondrial-targeted antioxidant not only decreased mtROS and MtD, it also significantly inhibited NLRP3 inflammasome activation and restored skeletal muscle atrophy. Finally, the PPAR-γ agonist protected against Ang II-induced muscle wasting by preventing MtD, oxidative stress, and NLRP3 inflammasome activation in vitro and in vivo. This work suggests a potential role of MtD/NLRP3 inflammasome pathway in the pathogenesis of Ang II-induced skeletal muscle wasting, and targeting the PPAR-γ/MtD/NLRP3 inflammasome axis may provide a therapeutic approach for muscle wasting.
Collapse
Affiliation(s)
- Yuqing Liu
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Xiao Bi
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Yumei Zhang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Yingdeng Wang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Wei Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China.
| |
Collapse
|
30
|
Leuchtmann AB, Handschin C. Pharmacological targeting of age-related changes in skeletal muscle tissue. Pharmacol Res 2020; 154:104191. [PMID: 30844535 PMCID: PMC7100900 DOI: 10.1016/j.phrs.2019.02.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/25/2022]
Abstract
Sarcopenia, the age-related loss of skeletal muscle mass and function, increases the risk of developing chronic diseases in older individuals and is a strong predictor of disability and death. Because of the ongoing demographic transition, age-related muscle weakness is responsible for an alarming and increasing contribution to health care costs in Western countries. Exercise-based interventions are most successful in preventing the decline in skeletal muscle mass and in preserving or ameliorating functional capacities with increasing age. However, other treatment options are still scarce. In this review, we explore currently applied nutritional and pharmacological approaches to mitigate age-related muscle wasting, and discuss potential future therapeutic avenues.
Collapse
Affiliation(s)
- Aurel B Leuchtmann
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056, Basel, Switzerland
| | - Christoph Handschin
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056, Basel, Switzerland.
| |
Collapse
|
31
|
Valle-Tenney R, Rebolledo D, Acuña MJ, Brandan E. HIF-hypoxia signaling in skeletal muscle physiology and fibrosis. J Cell Commun Signal 2020; 14:147-158. [PMID: 32088838 DOI: 10.1007/s12079-020-00553-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Hypoxia refers to the decrease in oxygen tension in the tissues, and the central effector of the hypoxic response is the transcription factor Hypoxia-Inducible Factor α (HIF1-α). Transient hypoxia in acute events, such as exercising or regeneration after damage, play an important role in skeletal muscle physiology and homeostasis. However, sustained activation of hypoxic signaling is a feature of skeletal muscle injury and disease, which can be a consequence of chronic damage but can also increase the severity of the pathology and worsen its outcome. Here, we review evidence that supports the idea that hypoxia and HIF-1α can contribute to the establishment of fibrosis in skeletal muscle through its crosstalk with other profibrotic factors, such as Transforming growth factor β (TGF-β), the induction of profibrotic cytokines expression, as is the case of Connective Tissue Growth Factor (CTGF/CCN2), or being the target of the Renin-angiotensin system (RAS).
Collapse
Affiliation(s)
- Roger Valle-Tenney
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago, Chile
| | - María José Acuña
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile. .,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Fundación Ciencia & Vida, Santiago, Chile. .,Department Cell and Molecular Biology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
32
|
Dehghani M, Mirzaie M, Farhadi P, Rezvani A. The Effect of ACE Inhibitor on the Quality of Life amongst Patients with Cancer Cachexia. Asian Pac J Cancer Prev 2020; 21:325-330. [PMID: 32102506 PMCID: PMC7332150 DOI: 10.31557/apjcp.2020.21.2.325] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Indexed: 11/25/2022] Open
Abstract
Background: ACEI (Angiotensin Converting Enzyme Inhibitors) inhibits tumor growth and development. Different mechanisms have been proposed for this matter, including the inhibition of enzymes that are involved in extracellular matrix degradation, matrix metalloproteinase (MMP) and etc. The present study was designed with the aim to investigate the effects of low dose ACEI on the Quality of Life (QoL) of non-hospitalized gastric cancer patients with cachexia. Materials and Methods: This study was a single-blinded randomized controlled clinical trial conducted in clinics affiliated with Shiraz University of Medical Sciences (SUMS). All participants were patients with gastric cancer in cancer cachexia step aged 40-80 years old who had referred to our clinics from October 2013 to April 2014. In the intervention group, patients were assigned to receive ACEI (Captopril) and the placebo group served as control and received placebo during the same time course. They were asked questions in order to fill out QLQ-C30 (Persian Version) questionnaire 3 times; baseline, 1 and 2 months after their first visit. Results: The mean age of patients was 60.55 ± 12.07 (range 31-80) years and the mean BMI of the patients was 17.21 ± 2.31. In the ACEI group, physical functioning and fatigue score changes were significant 1 and 2 months after treatment. The mean of fatigue score decreased significantly in the placebo group. Overall, global health status scores significantly increased in both groups, but other items of QoL did not change significantly. Conclusion: Overall, our results showed that Captopril does not have a significant positive effect on QoL of patients with cancer cachexia.
Collapse
Affiliation(s)
- Mehdi Dehghani
- Hematology Research Center, Department of Hematology and Medical Oncology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Mirzaie
- Department of Internal Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pouya Farhadi
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Rezvani
- Hematology Research Center, Department of Hematology and Medical Oncology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
33
|
Watanabe H, Enoki Y, Maruyama T. Sarcopenia in Chronic Kidney Disease: Factors, Mechanisms, and Therapeutic Interventions. Biol Pharm Bull 2020; 42:1437-1445. [PMID: 31474705 DOI: 10.1248/bpb.b19-00513] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic kidney disease (CKD), a chronic catabolic condition, is characterized by muscle wasting and decreased muscle endurance. Many insights into the molecular mechanisms of muscle wasting in CKD have been obtained. A persistent imbalance between protein degradation and synthesis in muscle causes muscle wasting. During muscle wasting, high levels of reactive oxygen species (ROS) and inflammatory cytokines are detected in muscle. These increased ROS and inflammatory cytokine levels induce the expression of myostatin. The myostatin binding to its receptor activin A receptor type IIB stimulates the expression of atrogenes such as atrogin-1 and muscle ring factor 1, members of the muscle-specific ubiquitin ligase family. Impaired mitochondrial function also contributes to reducing muscle endurance. The increased protein-bound uremic toxin, parathyroid hormone, glucocorticoid, and angiotensin II levels that are observed in CKD all have a negative effect on muscle mass and endurance. Among the protein-bound uremic toxins, indoxyl sulfate, an indole-containing compound has the potential to induce muscle atrophy by stimulating ROS-mediated myostatin and atrogenes expression. Indoxyl sulfate also impairs mitochondrial function. Some potential therapeutic approaches based on the muscle wasting mechanisms in CKD are currently in the testing stages.
Collapse
Affiliation(s)
- Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Enoki
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
34
|
Abstract
Modifications of lean mass are a frequent critical determinant in the pathophysiology and progression of heart failure (HF). Sarcopenia may be considered one of the most important causes of low physical performance and reduced cardiorespiratory fitness in older patients with HF. Sarcopenia is frequently misdiagnosed as cachexia. However, muscle wasting in HF has different pathogenetic features in sarcopenic and cachectic conditions. HF may induce sarcopenia through common pathogenetic pathways such as hormonal changes, malnutrition, and physical inactivity; mechanisms that influence each other. In the opposite way, sarcopenia may favor HF development by different mechanisms, including pathological ergoreflex. Paradoxically, sarcopenia is not associated with a sarcopenic cardiac muscle, but the cardiac muscle shows a hypertrophy which seems to be “not-functional.” First-line agents for the treatment of HF, physical activity and nutritional interventions, may offer a therapeutic advantage in sarcopenic patients irrespective of HF. Thus, sarcopenia is highly prevalent in patients with HF, contributing to its poor prognosis, and both conditions could benefit from common treatment strategies based on pharmacological, physical activity, and nutritional approaches.
Collapse
|
35
|
Lóry V, Balážová L, Kršková K, Horváthová Ľ, Olszanecki R, Suski M, Zórad Š. Obesity and aging affects skeletal muscle renin-angiotensin system and myosin heavy chain proportions in pre-diabetic Zucker rats. J Physiol Biochem 2019; 75:351-365. [PMID: 31197649 DOI: 10.1007/s13105-019-00689-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 05/26/2019] [Indexed: 10/26/2022]
Abstract
There is a gap in the knowledge regarding regulation of local renin-angiotensin system (RAS) in skeletal muscle during development of obesity and insulin resistance in vivo. This study evaluates the obesity- and age-related changes in the expression of local RAS components. Since RAS affects skeletal muscle remodelling, we also evaluated the muscle fibre type composition, defined by myosin heavy chain (MyHC) mRNAs and protein content. Gene expressions were determined by qPCR and/or Western blot analysis in musculus quadriceps of 3- and 8-month-old male obese Zucker rats and their lean controls. The enzymatic activity of aminopeptidase A (APA) was determined flourometrically. Activation of renin receptor (ReR)/promyelocytic leukaemia zinc finger (PLZF) negative feedback mechanism was observed in obesity. The expression of angiotensinogen and AT1 was downregulated by obesity, while neutral endopeptidase and AT2 expressions were upregulated in obese rats with aging. Skeletal muscle APA activity was decreased by obesity, which negatively correlated with the increased plasma APA activity and plasma cholesterol. The expression of angiotensin-converting enzyme (ACE) positively correlated with MyHC mRNAs characteristic for fast-twitch muscle fibres. The obesity- and age-related alterations in the expression of both classical and alternative RAS components suggest an onset of a new equilibrium between ACE/AngII/AT1 and ACE2/Ang1-7/Mas at lower level accompanied by increased renin/ReR/PLZF activation. Increased APA release from the skeletal muscle in obesity might contribute to increased plasma APA activity. There is a link between reduced ACE expression and altered muscle MyHC proportion in obesity and aging.
Collapse
Affiliation(s)
- Viktória Lóry
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava 4, Slovakia.
| | - Lucia Balážová
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava 4, Slovakia
| | - Katarína Kršková
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava 4, Slovakia
| | - Ľubica Horváthová
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava 4, Slovakia
| | - Rafal Olszanecki
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531, Krakow, Poland
| | - Maciej Suski
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531, Krakow, Poland
| | - Štefan Zórad
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava 4, Slovakia
| |
Collapse
|
36
|
Jafari‐Vayghan H, Saleh‐Ghadimi S, Maleki V, Moludi J, Alizadeh M. The effects of melatonin on neurohormonal regulation in cardiac cachexia: A mechanistic review. J Cell Biochem 2019; 120:16340-16351. [DOI: 10.1002/jcb.29151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Hamed Jafari‐Vayghan
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science Tabriz University of Medical Sciences Tabriz Iran
- Nutrition Research Center, Faculty of Nutrition and Food Sciences Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Sevda Saleh‐Ghadimi
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Vahid Maleki
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Jalal Moludi
- Department of Nutrition, Faculty of Nutrition Sciences and Food Technology Kermanshah University of Medical Sciences Kermanshah Iran
| | - Mohammad Alizadeh
- Department of Clinical Nutrition, Faculty of Nutrition and Food Science Tabriz University of Medical Sciences Tabriz Iran
- Nutrition Research Center, Faculty of Nutrition and Food Sciences Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
37
|
Chen P, Zhan Q, Bai Y, Huang X, Wang P, Pan Y, Li S, Fu S, Lai W, Zeng Q, Ren H, Xu D. Serum Peroxisome Proliferator-activated Receptor Gamma Coactivator-1α Related to Myocardial Energy Expenditure in Patients With Chronic Heart Failure. Am J Med Sci 2019; 357:205-212. [DOI: 10.1016/j.amjms.2018.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 11/29/2022]
|
38
|
Powers SK, Morton AB, Hyatt H, Hinkley MJ. The Renin-Angiotensin System and Skeletal Muscle. Exerc Sport Sci Rev 2018; 46:205-214. [PMID: 30001274 DOI: 10.1249/jes.0000000000000158] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The renin-angiotensin system (RAS) plays a key role in the control of blood pressure and fluid homeostasis. Emerging evidence also reveals that hyperactivity of the RAS contributes to skeletal muscle wasting. This review discusses the key role that the RAS plays in skeletal muscle wasting due to congestive heart failure, chronic kidney disease, and ventilator-induced diaphragmatic wasting.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | | | | | | |
Collapse
|
39
|
Modulation of the renin-angiotensin system in white adipose tissue and skeletal muscle: focus on exercise training. Clin Sci (Lond) 2018; 132:1487-1507. [PMID: 30037837 DOI: 10.1042/cs20180276] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/13/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022]
Abstract
Overactivation of the renin-angiotensin (Ang) system (RAS) increases the classical arm (Ang-converting enzyme (ACE)/Ang II/Ang type 1 receptor (AT1R)) to the detriment of the protective arm (ACE2/Ang 1-7/Mas receptor (MasR)). The components of the RAS are present locally in white adipose tissue (WAT) and skeletal muscle, which act co-operatively, through specific mediators, in response to pathophysiological changes. In WAT, up-regulation of the classical arm promotes lipogenesis and reduces lipolysis and adipogenesis, leading to adipocyte hypertrophy and lipid storage, which are related to insulin resistance and increased inflammation. In skeletal muscle, the classical arm promotes protein degradation and increases the inflammatory status and oxidative stress, leading to muscle wasting. Conversely, the protective arm plays a counter-regulatory role by opposing the effect of Ang II. The accumulation of adipose tissue and muscle mass loss is associated with a higher risk of morbidity and mortality, which could be related, in part, to overactivation of the RAS. On the other hand, exercise training (ExT) shifts the balance of the RAS towards the protective arm, promoting the inhibition of the classical arm in parallel with the stimulation of the protective arm. Thus, fat mobilization and maintenance of muscle mass and function are facilitated. However, the mechanisms underlying exercise-induced changes in the RAS remain unclear. In this review, we present the RAS as a key mechanism of WAT and skeletal muscle metabolic dysfunction. Furthermore, we discuss the interaction between the RAS and exercise and the possible underlying mechanisms of the health-related aspects of ExT.
Collapse
|
40
|
Kadoguchi T, Shimada K, Koide H, Miyazaki T, Shiozawa T, Takahashi S, Aikawa T, Ouchi S, Kitamura K, Sugita Y, Hamad AS, Kunimoto M, Sato-Okabayashi Y, Akita K, Isoda K, Daida H. Possible Role of NADPH Oxidase 4 in Angiotensin II-Induced Muscle Wasting in Mice. Front Physiol 2018; 9:340. [PMID: 29674975 PMCID: PMC5895660 DOI: 10.3389/fphys.2018.00340] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 03/20/2018] [Indexed: 12/25/2022] Open
Abstract
Background: Muscle wasting is a debilitating phenotype associated with chronic heart failure (CHF). We have previously demonstrated that angiotensin II (AII) directly induces muscle wasting in mice through the activation of NADPH oxidase (Nox). In this study, we tested the hypothesis that deficiency of NADPH oxidase 4 (Nox4), a major source of oxidative stress, ameliorates AII-induced muscle wasting through the regulation of redox balance. Methods and Results: Nox4 knockout (KO) and wild-type (WT) mice were used. At baseline, there were no differences in physical characteristics between the WT and KO mice. Saline (vehicle, V) or AII was infused via osmotic minipumps for 4 weeks, after which, the WT + AII mice showed significant increases in Nox activity and NOX4 protein compared with the WT + V mice, as well as decreases in body weight, gastrocnemius muscle weight, and myocyte cross-sectional area. These changes were significantly attenuated in the KO + AII mice (27 ± 1 vs. 31 ± 1 g, 385 ± 3 vs. 438 ± 13 mg, and 1,330 ± 30 vs. 2281 ± 150 μm2, respectively, all P < 0.05). The expression levels of phospho-Akt decreased, whereas those of muscle RING Finger-1 (MuRF-1) and MAFbx/atrogin-1 significantly increased in the WT + AII mice compared with the WT + V mice. Furthermore, nuclear factor erythroid-derived 2-like 2 (Nrf2) and the expression levels of Nrf2-regulated genes significantly decreased in the WT + AII mice compared with the WT + V mice. These changes were significantly attenuated in the KO + AII mice (P < 0.05). Conclusion: Nox4 deficiency attenuated AII-induced muscle wasting, partially through the regulation of Nrf2. The Nox4-Nrf2 axis may play an important role in the development of AII-induced muscle wasting.
Collapse
Affiliation(s)
- Tomoyasu Kadoguchi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kazunori Shimada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hiroshi Koide
- Laboratory of Molecular and Biochemical Research (Kyodo-ken), Research Support Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Tetsuro Miyazaki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Tomoyuki Shiozawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Shuhei Takahashi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Tatsuro Aikawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Shohei Ouchi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kenichi Kitamura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yurina Sugita
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Al Shahi Hamad
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Mitsuhiro Kunimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yayoi Sato-Okabayashi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Koji Akita
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kikuo Isoda
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
41
|
|
42
|
Sakuma K, Yamaguchi A. Drugs of Muscle Wasting and Their Therapeutic Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:463-481. [PMID: 30390265 DOI: 10.1007/978-981-13-1435-3_21] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Muscle wasting and weakness such as cachexia, atrophy, and sarcopenia are characterized by marked decreases in the protein content, myonuclear number, muscle fiber size, and muscle strength. This chapter focuses on the recent advances of pharmacological approach for attenuating muscle wasting.A myostatin-inhibiting approach is very intriguing to prevent sarcopenia but not muscular dystrophy in humans. Supplementation with ghrelin is also an important candidate to combat sarcopenia as well as cachexia. Treatment with soy isoflavone, trichostatin A (TSA), and cyclooxygenase 2 (Cox2) inhibitors seems to be effective modulators attenuating muscle wasting, although further systematic research is needed on this treatment in particular concerning side effects.
Collapse
Affiliation(s)
- Kunihiro Sakuma
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, Tokyo, Japan.
| | - Akihiko Yamaguchi
- Department of Physical Therapy, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| |
Collapse
|
43
|
Ábrigo J, Elorza AA, Riedel CA, Vilos C, Simon F, Cabrera D, Estrada L, Cabello-Verrugio C. Role of Oxidative Stress as Key Regulator of Muscle Wasting during Cachexia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2063179. [PMID: 29785242 PMCID: PMC5896211 DOI: 10.1155/2018/2063179] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
Abstract
Skeletal muscle atrophy is a pathological condition mainly characterized by a loss of muscular mass and the contractile capacity of the skeletal muscle as a consequence of muscular weakness and decreased force generation. Cachexia is defined as a pathological condition secondary to illness characterized by the progressive loss of muscle mass with or without loss of fat mass and with concomitant diminution of muscle strength. The molecular mechanisms involved in cachexia include oxidative stress, protein synthesis/degradation imbalance, autophagy deregulation, increased myonuclear apoptosis, and mitochondrial dysfunction. Oxidative stress is one of the most common mechanisms of cachexia caused by different factors. It results in increased ROS levels, increased oxidation-dependent protein modification, and decreased antioxidant system functions. In this review, we will describe the importance of oxidative stress in skeletal muscles, its sources, and how it can regulate protein synthesis/degradation imbalance, autophagy deregulation, increased myonuclear apoptosis, and mitochondrial dysfunction involved in cachexia.
Collapse
Affiliation(s)
- Johanna Ábrigo
- 1Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| | - Alvaro A. Elorza
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
- 3Centro de Investigaciones Biomédicas, Facultad de Ciencias Biológicas & Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Claudia A. Riedel
- 1Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| | - Cristian Vilos
- 4Laboratory of Nanomedicine and Targeted Delivery, Center for Integrative Medicine and Innovative Science, Faculty of Medicine, and Center for Bioinformatics and Integrative Biology, Faculty of Biological Sciences, Universidad Andres Bello, Santiago, Chile
- 5Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Felipe Simon
- 1Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| | - Daniel Cabrera
- 6Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- 7Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Lisbell Estrada
- 8Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Claudio Cabello-Verrugio
- 1Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| |
Collapse
|
44
|
Ábrigo J, Campos F, Simon F, Riedel C, Cabrera D, Vilos C, Cabello-Verrugio C. TGF-β requires the activation of canonical and non-canonical signalling pathways to induce skeletal muscle atrophy. Biol Chem 2017; 399:253-264. [DOI: 10.1515/hsz-2017-0217] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/03/2017] [Indexed: 12/20/2022]
Abstract
Abstract
The transforming growth factor type-beta (TGF-β) induces skeletal muscle atrophy characterised by a decrease in the fibre’s diameter and levels of myosin heavy chain (MHC), also as an increase of MuRF-1 expression. In addition, TGF-β induces muscle atrophy by a mechanism dependent on reactive oxygen species (ROS). TGF-β signals by activating both canonical Smad-dependent, and non-canonical signalling pathways such as ERK1/2, JNK1/2, and p38 MAPKs. However, the participation of canonical and non-canonical signalling pathways in the TGF-β atrophic effect on skeletal muscle is unknown. We evaluate the impact of Smad and MAPK signalling pathways on the TGF-β-induced atrophic effect in C2C12 myotubes. The results indicate that TGF-β activates Smad2/3, ERK1/2 and JNK1/2, but not p38 in myotubes. The pharmacological inhibition of Smad3, ERK1/2 and JNK1/2 activation completely abolished the atrophic effect of TGF-β. Finally, the inhibition of these canonical and non-canonical pathways did not decrease the ROS increment, while the inhibition of ROS production entirely abolished the phosphorylation of Smad3, ERK1/2 and JNK1/2. These results suggest that TGF-β requires Smad3, ERK1/2 and JNK1/2 activation to produce skeletal muscle atrophy. Moreover, the induction of ROS by TGF-β is an upstream event to canonical and non-canonical pathways.
Collapse
Affiliation(s)
- Johanna Ábrigo
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Fabian Campos
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Felipe Simon
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Claudia Riedel
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Daniel Cabrera
- Universidad Bernardo O Higgins, Facultad de Salud , Departamento de Ciencias Químicas y Biológicas , 8370993 Santiago , Chile
- Departamento de Gastroenterología, Facultad de Medicina , Pontificia Universidad Católica de Chile , 8331150 Santiago , Chile
| | - Cristian Vilos
- Laboratory of Nanomedicine and Targeted Delivery, Center for Integrative Medicine and Innovative Science, Faculty of Medicine, and Center for Bioinformatics and Integrative Biology, Faculty of Biological Sciences , Universidad Andres Bello , 8370146 Santiago , Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA) , Universidad de Santiago de Chile , 9170022 Santiago , Chile
| | - Claudio Cabello-Verrugio
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
- Laboratory of Muscle Pathology, Fragility and Aging , Departmento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina , Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
| |
Collapse
|
45
|
Becker BK, Wang H, Zucker IH. Central TrkB blockade attenuates ICV angiotensin II-hypertension and sympathetic nerve activity in male Sprague-Dawley rats. Auton Neurosci 2017; 205:77-86. [PMID: 28549782 DOI: 10.1016/j.autneu.2017.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/17/2017] [Accepted: 05/17/2017] [Indexed: 02/07/2023]
Abstract
Increased sympathetic nerve activity and the activation of the central renin-angiotensin system are commonly associated with cardiovascular disease states such as hypertension and heart failure, yet the precise mechanisms contributing to the long-term maintenance of this sympatho-excitation are incompletely understood. Due to the established physiological role of neurotrophins contributing toward neuroplasticity and neuronal excitability along with recent evidence linking the renin-angiotensin system and brain-derived neurotrophic factor (BDNF) along with its receptor (TrkB), it is likely the two systems interact to promote sympatho-excitation during cardiovascular disease. However, this interaction has not yet been fully demonstrated, in vivo. Thus, we hypothesized that central angiotensin II (Ang II) treatment will evoke a sympatho-excitatory state mediated through the actions of BDNF/TrkB. We infused Ang II (20ng/min) into the right lateral ventricle of male Sprague-Dawley rats for twelve days with or without the TrkB receptor antagonist, ANA-12 (50ng/h). We found that ICV infusion of Ang II increased mean arterial pressure (+40.4mmHg), increased renal sympathetic nerve activity (+19.4% max activity), and induced baroreflex dysfunction relative to vehicle. Co-infusion of ANA-12 attenuated the increase in blood pressure (-20.6mmHg) and prevented the increase in renal sympathetic nerve activity (-22.2% max) and baroreflex dysfunction relative to Ang II alone. Ang II increased thirst and decreased food consumption, and Ang II+ANA-12 augmented the thirst response while attenuating the decrease in food consumption. We conclude that TrkB signaling is a mediator of the long-term blood pressure and sympathetic nerve activity responses to central Ang II activity. These findings demonstrate the involvement of neurotrophins such as BDNF in promoting Ang II-induced autonomic dysfunction and further implicate TrkB signaling in modulating presympathetic autonomic neurons during cardiovascular disease.
Collapse
Affiliation(s)
- Bryan K Becker
- Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hanjun Wang
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, USA.; Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Irving H Zucker
- Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA..
| |
Collapse
|
46
|
Leitner LM, Wilson RJ, Yan Z, Gödecke A. Reactive Oxygen Species/Nitric Oxide Mediated Inter-Organ Communication in Skeletal Muscle Wasting Diseases. Antioxid Redox Signal 2017; 26:700-717. [PMID: 27835923 PMCID: PMC5421600 DOI: 10.1089/ars.2016.6942] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Cachexia is defined as a complex metabolic syndrome that is associated with underlying illness and a loss of muscle with or without loss of fat mass. This disease is associated with a high incidence with chronic diseases such as heart failure, cancer, chronic obstructive pulmonary disease (COPD), and acquired immunodeficiency syndrome (AIDS), among others. Since there is currently no effective treatment available, cachectic patients have a poor prognosis. Elucidation of the underlying mechanisms is, therefore, an important medical task. Recent Advances: There is accumulating evidence that the diseased organs such as heart, lung, kidney, or cancer tissue secrete soluble factors, including Angiotensin II, myostatin (growth differentiation factor 8 [GDF8]), GDF11, tumor growth factor beta (TGFβ), which act on skeletal muscle. There, they induce a set of genes called atrogenes, which, among others, induce the ubiquitin-proteasome system, leading to protein degradation. Moreover, elevated reactive oxygen species (ROS) levels due to modulation of NADPH oxidases (Nox) and mitochondrial function contribute to disease progression, which is characterized by loss of muscle mass, exercise resistance, and frailty. CRITICAL ISSUES Although substantial progress was achieved to elucidate the pathophysiology of cachexia, effectice therapeutic strategies are urgently needed. FUTURE DIRECTIONS With the identification of key components of the aberrant inter-organ communication leading to cachexia, studies in mice and men to inhibit ROS formation, induction of anti-oxidative superoxide dismutases, and upregulation of muscular nitric oxide (NO) formation either by pharmacological tools or by exercise are promising approaches to reduce the extent of skeletal muscle wasting. Antioxid. Redox Signal. 26, 700-717.
Collapse
Affiliation(s)
- Lucia M Leitner
- 1 Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität Düsseldorf, Universitätsklinikum , Düsseldorf, Germany
| | - Rebecca J Wilson
- 2 Department of Medicine-Cardiovascular Medicine, University of Virginia , Charlottesville, Virginia
| | - Zhen Yan
- 2 Department of Medicine-Cardiovascular Medicine, University of Virginia , Charlottesville, Virginia.,3 Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia , Charlottesville, Virginia
| | - Axel Gödecke
- 1 Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität Düsseldorf, Universitätsklinikum , Düsseldorf, Germany
| |
Collapse
|
47
|
Saitoh M, Ishida J, Doehner W, von Haehling S, Anker MS, Coats AJS, Anker SD, Springer J. Sarcopenia, cachexia, and muscle performance in heart failure: Review update 2016. Int J Cardiol 2017; 238:5-11. [PMID: 28427849 DOI: 10.1016/j.ijcard.2017.03.155] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023]
Abstract
Cachexia in the context of heart failure (HF) has been termed cardiac cachexia, and represents a progressive involuntary weight loss. Cachexia is mainly the result of an imbalance in the homeostasis of muscle protein synthesis and degradation due to a lower activity of protein synthesis pathways and an over-activation of protein degradation. In addition, muscle wasting leads to of impaired functional capacity, even after adjusting for clinical relevant variables in patients with HF. However, there is no sufficient therapeutic strategy in muscle wasting in HF patients and very few studies in animal models. Exercise training represents a promising intervention that can prevent or even reverse the process of muscle wasting, and worsening the muscle function and performance in HF with muscle wasting and cachexia. The pathological mechanisms and effective therapeutic approach of cardiac cachexia remain uncertain, because of the difficulty to establish animal cardiac cachexia models, thus novel animal models are warranted. Furthermore, the use of improved animal models will lead to a better understanding of the pathways that modulate muscle wasting and therapeutics of muscle wasting of cardiac cachexia.
Collapse
Affiliation(s)
- Masakazu Saitoh
- Institute of Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Junichi Ishida
- Institute of Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfram Doehner
- Charité - Campus Virchow (CVK), Center for Stroke Research, Berlin, Germany
| | - Stephan von Haehling
- Institute of Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Markus S Anker
- Charité - Campus Benjamin Franklin (CBF), Department of Cardiology, Berlin, Germany Charité - Campus Virchow (CVK), Center for Stroke Research, Berlin, Germany
| | | | - Stefan D Anker
- Institute of Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Jochen Springer
- Institute of Innovative Clinical Trials, Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
48
|
Shen C, Zhou J, Wang X, Yu XY, Liang C, Liu B, Pan X, Zhao Q, Song JL, Wang J, Bao M, Wu C, Li Y, Song YH. Angiotensin-II-induced Muscle Wasting is Mediated by 25-Hydroxycholesterol via GSK3β Signaling Pathway. EBioMedicine 2017; 16:238-250. [PMID: 28161398 PMCID: PMC5474518 DOI: 10.1016/j.ebiom.2017.01.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 01/20/2017] [Accepted: 01/27/2017] [Indexed: 12/14/2022] Open
Abstract
While angiotensin II (ang II) has been implicated in the pathogenesis of cardiac cachexia (CC), the molecules that mediate ang II's wasting effect have not been identified. It is known TNF-α level is increased in patients with CC, and TNF-α release is triggered by ang II. We therefore hypothesized that ang II induced muscle wasting is mediated by TNF-α. Ang II infusion led to skeletal muscle wasting in wild type (WT) but not in TNF alpha type 1 receptor knockout (TNFR1KO) mice, suggesting that ang II induced muscle loss is mediated by TNF-α through its type 1 receptor. Microarray analysis identified cholesterol 25-hydroxylase (Ch25h) as the down stream target of TNF-α. Intraperitoneal injection of 25-hydroxycholesterol (25-OHC), the product of Ch25h, resulted in muscle loss in C57BL/6 mice, accompanied by increased expression of atrogin-1, MuRF1 and suppression of IGF-1/Akt signaling pathway. The identification of 25-OHC as an inducer of muscle wasting has implications for the development of specific treatment strategies in preventing muscle loss. Ang II induced muscle wasting is mediated by TNF-α, which in turn up regulates Ch25h Knockout of TNFR1 inhibits the production of 25-OHC and blocks ang II induced muscle loss in mice 25-OHC injection induces muscle wasting in mice by activating GSK3β A GSK3β inhibitor blocks ang II induced muscle atrophy, which paves the way for targeted therapy to treat muscle wasting
Cardiac cachexia (CC), a condition characterized by weight loss and muscle wasting, is a serious complication that occurs in patients with chronic heart failure. This condition impairs patient's daily physical activity and their quality of life. Specific therapy for CC is currently unavailable because the pathogenesis remains unknown. Previous studies have identified angiotensin II (ang II) as an important mediator of CC. We now report a previously unrecognized role of 25-hydroxycholesterol (25-OHC) in mediating ang II induced muscle loss. The identification of 25-OHC as a muscle wasting inducer has implications for the development of therapeutic intervention in preserving muscle mass.
Collapse
Affiliation(s)
- Congcong Shen
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Jin Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, PR China.
| | - Xiaoxiao Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Xi-Yong Yu
- Guangdong Cardiovascular Institute, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Chun Liang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, No. 415, Fengyang Road, Shanghai, PR China
| | - Bin Liu
- Cardiovascular Disease Center, The First Hospital of Ji Lin University, Changchun, Jilin 130021, PR China
| | - Xiangbin Pan
- Department of Cardiac Surgery, Fuwai Hospital, PR China
| | - Qiong Zhao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Jenny Lee Song
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Jiajun Wang
- Department of Gynecology, The Affiliated Maternity and Child Health Hospital of Nanjing Medical University, Wuxi, PR China
| | - Meiyu Bao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Chaofan Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Yangxin Li
- The Department of Cardiovascular Surgery of the First Affiliated Hospital and the Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215123, PR China.
| | - Yao-Hua Song
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China.
| |
Collapse
|
49
|
Xiao C, Wu Q, Zhang J, Xie Y, Cai W, Tan J. Antidiabetic activity of Ganoderma lucidum polysaccharides F31 down-regulated hepatic glucose regulatory enzymes in diabetic mice. JOURNAL OF ETHNOPHARMACOLOGY 2017; 196:47-57. [PMID: 27902927 DOI: 10.1016/j.jep.2016.11.044] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 11/23/2016] [Accepted: 11/26/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ganoderma lucidum (Lin Zhi) has been used to treat diabetes in Chinese folk for centuries. Our laboratory previously demonstrated that Ganoderma lucidum polysaccharides (GLPs) had hypoglycemic effects in diabetic mice. Our aim was to identify the main bioactives in GLPs and corresponding mechanism of action. MATERIALS AND METHODS Four polysaccharide-enriched fraction were isolated from GLPs and the antidiabetic activities were evaluated by type 2 diabetic mice. Fasting serum glucose (FSG), fasting serum insulin (FSI) and epididymal fat/BW ratio were measured at the end of the experiment. In liver, the mRNA levels of hepatic glucose regulatory enzymes were determined by quantitative polymerase chain reaction (qPCR) and the protein levels of phospho-AMP-activated protein kinase (p-AMPK)/AMPK were determined by western blotting test. In epididymal fat tissue, the mRNA and protein levels GLUT4, resistin, fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC1) were determined by qPCR and immuno-histochemistry. The structure of polysaccharide F31 was obtained from GPC, FTIR NMR and GC-MS spectroscopy, RESULTS: F31 significantly decreased FSG (P<0.05), FSI and epididymal fat/BW ratio (P<0.01). In liver, F31 decreased the mRNA levels of hepatic glucose regulatory enzymes, and up-regulated the ratio of phospho-AMP-activated protein kinase (p-AMPK)/AMPK. In epididymal fat tissue, F31 increased the mRNA levels of GLUT4 but decreased fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC1) and resistin. Immuno-histochemistry results revealed F31 increased the protein levels of GLUT4 and decreased resistin. CONCLUSION Data suggested that the main bioactives in GLPs was F31, which was determined to be a β-heteropolysaccharide with the weight-average molecular weight of 15.9kDa. The possible action mechanism of F31 may be associated with down-regulation of the hepatic glucose regulated enzyme mRNA levels via AMPK activation, improvement of insulin resistance and decrease of epididymal fat/BW ratio. These results strongly suggest that F31 has antidiabetic potential.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Acetyl-CoA Carboxylase/genetics
- Acetyl-CoA Carboxylase/metabolism
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Animals
- Blood Glucose/analysis
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Down-Regulation
- Fasting/blood
- Fatty Acid Synthase, Type I/genetics
- Fatty Acid Synthase, Type I/metabolism
- Fruiting Bodies, Fungal
- Fungal Polysaccharides/pharmacology
- Fungal Polysaccharides/therapeutic use
- Ganoderma
- Glucose Transporter Type 4/genetics
- Glucose Transporter Type 4/metabolism
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin/blood
- Liver/drug effects
- Liver/metabolism
- Male
- Mice, Inbred C57BL
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Chun Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Xianlie Central Road 100, Guangzhou 510070, China.
| | - Qingping Wu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Xianlie Central Road 100, Guangzhou 510070, China.
| | - Jumei Zhang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Xianlie Central Road 100, Guangzhou 510070, China.
| | - Yizhen Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Xianlie Central Road 100, Guangzhou 510070, China.
| | - Wen Cai
- Department of Toxicology, Center for Disease Control and Prevention of Guangdong Province, Guangzhou 510020, China.
| | - Jianbin Tan
- Department of Toxicology, Center for Disease Control and Prevention of Guangdong Province, Guangzhou 510020, China.
| |
Collapse
|
50
|
The mechanisms of breathlessness in heart failure as the basis of therapy. Curr Opin Support Palliat Care 2016; 10:32-5. [PMID: 26716391 DOI: 10.1097/spc.0000000000000181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW The review provides an overview of recent understanding in relation to the mechanisms relating to skeletal muscle and the sympathetic nervous system, and therapies for breathlessness which target these mechanisms. These are set in the context of established knowledge in this field. RECENT FINDINGS Despite strong evidence to support exercise training programmes, and recommendations in international guidelines, programmes are implemented poorly. Electrical stimulation appears to be a way of exercising people too frail to undertake a full exercise programme. There is evidence to support the use of opioids for breathlessness in other conditions, but as yet the evidence in chronic heart failure is mixed. SUMMARY Previous work in relation to the role of skeletal muscle and sympathetic nervous system has set the scene for targeted therapies for the relief of breathlessness in people with heart failure.
Collapse
|