1
|
Hoekstra S, King JA, Fenton J, Kirk N, Willis SA, Phillips SM, Webborn N, Tolfrey K, Bosch JDVD, Goosey‐Tolfrey VL. The effect of home-based neuromuscular electrical stimulation-resistance training and protein supplementation on lean mass in persons with spinal cord injury: A pilot study. Physiol Rep 2024; 12:e70073. [PMID: 39358836 PMCID: PMC11446856 DOI: 10.14814/phy2.70073] [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: 08/26/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
In persons with a spinal cord injury (SCI), resistance training using neuromuscular electrical stimulation (NMES-RT) increases lean mass in the lower limbs. However, whether protein supplementation in conjunction with NMES-RT further enhances this training effect is unknown. In this randomized controlled pilot trial, 15 individuals with chronic SCI engaged in 3 times/week NMES-RT, with (NMES+PRO, n = 8) or without protein supplementation (NMES, n = 7), for 12 weeks. Before and after the intervention, whole body and regional body composition (DXA) and fasting glucose and insulin concentrations were assessed in plasma. Adherence to the intervention components was ≥96%. Thigh lean mass was increased to a greater extent after NMES+PRO compared to NMES (0.3 (0.2, 0.4) kg; p < 0.001). Furthermore, fasting insulin concentration and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) were decreased similarly in both groups (fasting insulin: 1 [-9, 11] pmol∙L-1; HOMA-IR: 0.1 [-0.3, 0.5] AU; both p ≥ 0.617). Twelve weeks of home-based NMES-RT increased thigh lean mass, an effect that was potentiated by protein supplementation. In combination with the excellent adherence and apparent improvement in cardiometabolic health outcomes, these findings support further investigation through a full-scale randomized controlled trial.
Collapse
Affiliation(s)
- Sven Hoekstra
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
- Department of Exercise and Sport ScienceSt. Mary's UniversitySan AntonioTexasUSA
- Department of Rehabilitation MedicineUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - James A. King
- School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
- National Institute for Health and Care Research (NIHR) Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of LeicesterLeicesterUK
| | - Jordan Fenton
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
- School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
| | - Natasha Kirk
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
- School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
| | - Scott A. Willis
- School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
- National Institute for Health and Care Research (NIHR) Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of LeicesterLeicesterUK
| | | | - Nick Webborn
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
| | - Keith Tolfrey
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
- School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
| | | | - Vicky L. Goosey‐Tolfrey
- Peter Harrison Centre for Disability Sport, School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
- School of Sport, Exercise and Health SciencesLoughborough UniversityLeicestershireUK
| |
Collapse
|
2
|
da Silva RP, Costa DM, da Cruz-Filho J, Santos TDO, Dos Anjos-Santos HC, Vasconcelos ABS, Heck LC, Kettelhut ÍDC, Navegantes LC, Dos Santos JR, de Souza PRM, Badauê-Passos D, Mecawi AS, DeSantana JM, Lustrino D. Reduced sympathetic activity is associated with the development of pain and muscle atrophy in a female rat model of fibromyalgia. Physiol Behav 2024; 281:114575. [PMID: 38692384 DOI: 10.1016/j.physbeh.2024.114575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/16/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
Fibromyalgia (FM) is characterized by chronic widespread musculoskeletal pain accompanied by fatigue and muscle atrophy. Although its etiology is not known, studies have shown that FM patients exhibit altered function of the sympathetic nervous system (SNS), which regulates nociception and muscle plasticity. Nevertheless, the precise SNS-mediated mechanisms governing hyperalgesia and skeletal muscle atrophy in FM remain unclear. Thus, we employed two distinct FM-like pain models, involving intramuscular injections of acidic saline (pH 4.0) or carrageenan in prepubertal female rats, and evaluated the catecholamine content, adrenergic signaling and overall muscle proteolysis. Subsequently, we assessed the contribution of the SNS to the development of hyperalgesia and muscle atrophy in acidic saline-injected rats treated with clenbuterol (a selective β2-adrenergic receptor agonist) and in animals maintained under baseline conditions and subjected to epinephrine depletion through adrenodemedullation (ADM). Seven days after inducing an FM-like model with acidic saline or carrageenan, we observed widespread mechanical hyperalgesia along with loss of strength and/or muscle mass. These changes were associated with reduced catecholamine content, suggesting a common underlying mechanism. Notably, treatment with a β2-agonist alleviated hyperalgesia and prevented muscle atrophy in acidic saline-induced FM-like pain, while epinephrine depletion induced mechanical hyperalgesia and increased muscle proteolysis in animals under baseline conditions. Together, the results suggest that reduced sympathetic activity is involved in the development of pain and muscle atrophy in the murine model of FM analyzed.
Collapse
Affiliation(s)
- Raquel Prado da Silva
- Laboratory of Basic and Behavioral Neuroendocrinology (LANBAC), Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Daniely Messias Costa
- Laboratory of Basic and Behavioral Neuroendocrinology (LANBAC), Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - João da Cruz-Filho
- Laboratory of Basic and Behavioral Neuroendocrinology (LANBAC), Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Tatiane de Oliveira Santos
- Laboratory of Basic and Behavioral Neuroendocrinology (LANBAC), Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Hevely Catharine Dos Anjos-Santos
- Laboratory of Basic and Behavioral Neuroendocrinology (LANBAC), Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Alan Bruno Silva Vasconcelos
- Laboratory of Basic and Behavioral Neuroendocrinology (LANBAC), Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Lilian Carmo Heck
- Department of Physiology and Biochemistry & Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ísis do Carmo Kettelhut
- Department of Physiology and Biochemistry & Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luiz Carlos Navegantes
- Department of Physiology and Biochemistry & Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - José Ronaldo Dos Santos
- Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | - Daniel Badauê-Passos
- Laboratory of Basic and Behavioral Neuroendocrinology (LANBAC), Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - André Souza Mecawi
- Department of Biophysics, São Paulo Medical School, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Josimari Melo DeSantana
- Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduate Program in Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Danilo Lustrino
- Laboratory of Basic and Behavioral Neuroendocrinology (LANBAC), Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduate Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil.
| |
Collapse
|
3
|
Delbono O, Wang Z, Messi ML. Brainstem noradrenergic neurons: Identifying a hub at the intersection of cognition, motility, and skeletal muscle regulation. Acta Physiol (Oxf) 2022; 236:e13887. [PMID: 36073023 PMCID: PMC9588743 DOI: 10.1111/apha.13887] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 01/29/2023]
Abstract
Brainstem noradrenergic neuron clusters form a node integrating efferents projecting to distinct areas such as those regulating cognition and skeletal muscle structure and function, and receive dissimilar afferents through established circuits to coordinate organismal responses to internal and environmental challenges. Genetic lineage tracing shows the remarkable heterogeneity of brainstem noradrenergic neurons, which may explain their varied functions. They project to the locus coeruleus, the primary source of noradrenaline in the brain, which supports learning and cognition. They also project to pre-ganglionic neurons, which lie within the spinal cord and form synapses onto post-ganglionic neurons. The synapse between descending brainstem noradrenergic neurons and pre-ganglionic spinal neurons, and these in turn with post-ganglionic noradrenergic neurons located at the paravertebral sympathetic ganglia, support an anatomical hierarchy that regulates skeletal muscle innervation, neuromuscular transmission, and muscle trophism. Whether any noradrenergic neuron subpopulation is more susceptible to damaged protein deposit and death with ageing and neurodegeneration is a relevant question that answer will help us to detect neurodegeneration at an early stage, establish prognosis, and anticipate disease progression. Loss of muscle mass and strength with ageing, termed sarcopenia, may predict impaired cognition with ageing and neurodegeneration and establish an early time to start interventions aimed at reducing central noradrenergic neurons hyperactivity. Complex multidisciplinary approaches, including genetic tracing, specific circuit labelling, optogenetics and chemogenetics, electrophysiology, and single-cell transcriptomics and proteomics, are required to test this hypothesis pre-clinical.
Collapse
Affiliation(s)
- Osvaldo Delbono
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Zhong‐Min Wang
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - María Laura Messi
- Department of Internal MedicineSection on Gerontology and Geriatric Medicine. Wake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
4
|
Franzoso M, Dokshokova L, Vitiello L, Zaglia T, Mongillo M. Tuning the Consonance of Microscopic Neuro-Cardiac Interactions Allows the Heart Beats to Play Countless Genres. Front Physiol 2022; 13:841740. [PMID: 35273522 PMCID: PMC8902305 DOI: 10.3389/fphys.2022.841740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Different from skeletal muscle, the heart autonomously generates rhythmic contraction independently from neuronal inputs. However, speed and strength of the heartbeats are continuously modulated by environmental, physical or emotional inputs, delivered by cardiac innervating sympathetic neurons, which tune cardiomyocyte (CM) function, through activation of β-adrenoceptors (β-ARs). Given the centrality of such mechanism in heart regulation, β-AR signaling has been subject of intense research, which has reconciled the molecular details of the transduction pathway and the fine architecture of cAMP signaling in subcellular nanodomains, with its final effects on CM function. The importance of mechanisms keeping the elements of β-AR/cAMP signaling in good order emerges in pathology, when the loss of proper organization of the transduction pathway leads to detuned β-AR/cAMP signaling, with detrimental consequences on CM function. Despite the compelling advancements in decoding cardiac β-AR/cAMP signaling, most discoveries on the subject were obtained in isolated cells, somehow neglecting that complexity may encompass the means in which receptors are activated in the intact heart. Here, we outline a set of data indicating that, in the context of the whole myocardium, the heart orchestra (CMs) is directed by a closely interacting and continuously attentive conductor, represented by SNs. After a roundup of literature on CM cAMP regulation, we focus on the unexpected complexity and roles of cardiac sympathetic innervation, and present the recently discovered Neuro-Cardiac Junction, as the election site of "SN-CM" interaction. We further discuss how neuro-cardiac communication is based on the combination of extra- and intra-cellular signaling micro/nano-domains, implicating neuronal neurotransmitter exocytosis, β-ARs and elements of cAMP homeostasis in CMs, and speculate on how their dysregulation may reflect on dysfunctional neurogenic control of the heart in pathology.
Collapse
Affiliation(s)
- Mauro Franzoso
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lolita Dokshokova
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
5
|
Saleh AA, Alhotan RA, Alharthi AS, Nassef E, Kassab MA, Farrag FA, Hendam BM, Abumnadour MMA, Shukry M. Insight View on the Role of in Ovo Feeding of Clenbuterol on Hatched Chicks: Hatchability, Growth Efficiency, Serum Metabolic Profile, Muscle, and Lipid-Related Markers. Animals (Basel) 2021; 11:ani11082429. [PMID: 34438887 PMCID: PMC8388663 DOI: 10.3390/ani11082429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary This study examined the effects of ovo injection of clenbuterol on fat deposition and growth performance in chickens, which is prejudicial to poultry consumers and muscle growth-related genes, egg hatchability, and fertility. The achieved result showed a definite effect of clenbuterol on body gain and hatchability. It decreased fat deposition and upregulation of muscle growth-related gene expressions accompanied by modulation of fatty and amino acid composition, reflecting a new insight into the intracellular pathways of clenbuterol supplementation on chicks. Abstract The present study aimed to assess the in ovo administration of clenbuterol on chick fertility, growth performance, muscle growth, myogenic gene expression, fatty acid, amino acid profile, intestinal morphology, and hepatic lipid-related gene expressions. In this study, 750 healthy fertile eggs from the local chicken breed Dokki-4 strain were analyzed. Fertile eggs were randomly divided into five experimental groups (150 eggs/3 replicates for each group). On day 14 of incubation, in addition to the control group, four other groups were established where 0.5 mL of worm saline (30 °C) was injected into the second group of eggs. In the third, fourth, and fifth groups, 0.5 mL of worm saline (30 °C), 0.9% of NaCl, and 10, 15, and 20 ppm of clenbuterol were injected into the eggs. Results suggested that clenbuterol increased growth efficiency up to 12 weeks of age, especially at 15 ppm, followed by 10 ppm, decreased abdominal body fat mass, and improved hatchability (p < 0.01). Clenbuterol also modulated saturated fatty acid levels in the breast muscles and improved essential amino acids when administered at 10 and 15 ppm. Additionally, clenbuterol at 15 ppm significantly decreased myostatin gene expression (p < 0.01) and considerably increased IGF1r and IGF-binding protein (IGFBP) expression. Clenbuterol administration led to a significant upregulation of hepatic PPARα, growth hormone receptor, and Lipoprotein lipase (LPL) mRNA expression with a marked decrease in fatty acid synthase (FAS) and sterol regulatory element-binding protein 1 (SREBP-1c) expression. In conclusion, the current study revealed that in ovo injection of clenbuterol showed positive effects on the growth of hatched chicks through reduced abdominal fat deposition, improved intestinal morphology, and modulation of hepatic gene expressions in myogenesis, lipogenesis, and lipolysis.
Collapse
Affiliation(s)
- Ahmed A. Saleh
- Department of Poultry Production, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
- Correspondence: (A.A.S.); (M.S.)
| | - Rashed A. Alhotan
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (R.A.A.); (A.S.A.)
| | - Abdulrahman S. Alharthi
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia; (R.A.A.); (A.S.A.)
| | - Eldsokey Nassef
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Mohamed A. Kassab
- Department of Histology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Foad A. Farrag
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Basma M. Hendam
- Genetics and Genetic Engineering, Department of Husbandry and Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohamed M. A. Abumnadour
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Alexandria University, Edfina 22756, Egypt;
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
- Correspondence: (A.A.S.); (M.S.)
| |
Collapse
|
6
|
Blears E, Ross E, Ogunbileje JO, Porter C, Murton AJ. The impact of catecholamines on skeletal muscle following massive burns: Friend or foe? Burns 2021; 47:756-764. [PMID: 33568281 DOI: 10.1016/j.burns.2021.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
Profound skeletal muscle wasting in the setting of total body hypermetabolism is a defining characteristic of massive burns, compromising the patient's recovery and necessitating a protracted period of rehabilitation. In recent years, the prolonged use of the non-selective beta-blocker, propranolol, has gained prominence as an effective tool to assist with suppressing epinephrine-dependent burn-induced hypermetabolism and by extension, blunting muscle catabolism. However, synthetic β-adrenergic agonists, such as clenbuterol, are widely associated with the promotion of muscle growth in both animals and humans. Moreover, experimental adrenodemedullation is known to result in muscle catabolism. Therefore, the blunting of muscle β-adrenergic signaling via the use of propranolol would be expected to negatively impair muscle protein homeostasis. This review explores these paradoxical observations and identifies the manner by which propranolol is thought to exert its anti-catabolic effects in burn patients. Moreover, we identify potential avenues by which the use of beta-blocker therapy in the treatment of massive burns could potentially be further refined to promote the recovery of muscle mass in these critically ill patients while continuing to ameliorate total body hypermetabolism.
Collapse
Affiliation(s)
- Elizabeth Blears
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA; Department of Surgery, Allegheny Health Network, Pittsburgh, PA, USA
| | - Evan Ross
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - John O Ogunbileje
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Craig Porter
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA; Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Andrew J Murton
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center of Aging, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
7
|
Rodrigues ACZ, Wang ZM, Messi ML, Bonilla HJ, Liu L, Freeman WM, Delbono O. Heart and neural crest derivative 2-induced preservation of sympathetic neurons attenuates sarcopenia with aging. J Cachexia Sarcopenia Muscle 2021; 12:91-108. [PMID: 33258279 PMCID: PMC7890150 DOI: 10.1002/jcsm.12644] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 10/02/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Sarcopenia, or age-dependent decline in muscle force and power, impairs mobility, increasing the risk of falls, institutionalization, co-morbidity, and premature death. The discovery of adrenoceptors, which mediate the effects of the sympathetic nervous system (SNS) neurotransmitter norepinephrine on specific tissues, sparked the development of sympathomimetics that have profound influence on skeletal muscle mass. However, chronic administration has serious side effects that preclude their use for muscle-wasting conditions. Interventions that can adjust neurotransmitter release to changing physiological demands depend on understanding how the SNS affects neuromuscular transmission, muscle motor innervation, and muscle mass. METHODS We examined age-dependent expression of the heart and neural crest derivative 2 (Hand2), a critical transcription factor for SN maintenance, and we tested the possibility that inducing its expression exclusively in sympathetic neurons (SN) will prevent (i) motor denervation, (ii) impaired neuromuscular junction (NMJ) transmission, and (iii) loss of muscle mass and function in old mice. To test this hypothesis, we delivered a viral vector carrying Hand2 expression or an empty vector exclusively in SNs by vein injection in 16-month-old C57BL/6 mice that were sacrificed 6 months later. Techniques include RNA-sequencing, real-time PCR, genomic DNA methylation, viral vector construct, tissue immunohistochemistry, immunoblot, confocal microscopy, electrophysiology, and in vivo mouse physical performance. RESULTS Hand2 expression declines throughout life, but inducing its expression increased (i) the number and size of SNs, (ii) muscle sympathetic innervation, (iii) muscle weight and force and whole-body strength, (iv) myofiber size but not muscle fibre-type composition, (v) NMJ transmission and nerve-evoked muscle force, and (vi) motor innervation in old mice. Additionally, the SN controls a set of genes to reduce inflammation and to promote transcription factor activity, cell signalling, and synapse in the skeletal muscle. Hand2 DNA methylation may contribute, at least partially, to gene silencing. CONCLUSIONS Selective expression of Hand2 in the mouse SNs from middle age through old age increases muscle mass and force by (i) regulating skeletal muscle sympathetic and motor innervation; (ii) improving acetylcholine receptor stability and NMJ transmission; (iii) preventing inflammation and myofibrillar protein degradation; (iv) increasing autophagy; and (v) probably enhancing protein synthesis.
Collapse
Affiliation(s)
- Anna Carolina Zaia Rodrigues
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Zhong-Min Wang
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - María Laura Messi
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Henry Jacob Bonilla
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Liang Liu
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Osvaldo Delbono
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
8
|
Azevedo Voltarelli V, Coronado M, Gonçalves Fernandes L, Cruz Campos J, Jannig PR, Batista Ferreira JC, Fajardo G, Chakur Brum P, Bernstein D. β 2-Adrenergic Signaling Modulates Mitochondrial Function and Morphology in Skeletal Muscle in Response to Aerobic Exercise. Cells 2021; 10:cells10010146. [PMID: 33450889 PMCID: PMC7828343 DOI: 10.3390/cells10010146] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/28/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
The molecular mechanisms underlying skeletal muscle mitochondrial adaptations induced by aerobic exercise (AE) are not fully understood. We have previously shown that AE induces mitochondrial adaptations in cardiac muscle, mediated by sympathetic stimulation. Since direct sympathetic innervation of neuromuscular junctions influences skeletal muscle homeostasis, we tested the hypothesis that β2-adrenergic receptor (β2-AR)-mediated sympathetic activation induces mitochondrial adaptations to AE in skeletal muscle. Male FVB mice were subjected to a single bout of AE on a treadmill (80% Vmax, 60 min) under β2-AR blockade with ICI 118,551 (ICI) or vehicle, and parameters of mitochondrial function and morphology/dynamics were evaluated. An acute bout of AE significantly increased maximal mitochondrial respiration in tibialis anterior (TA) isolated fiber bundles, which was prevented by β2-AR blockade. This increased mitochondrial function after AE was accompanied by a change in mitochondrial morphology towards fusion, associated with increased Mfn1 protein expression and activity. β2-AR blockade fully prevented the increase in Mfn1 activity and reduced mitochondrial elongation. To determine the mechanisms involved in mitochondrial modulation by β2-AR activation in skeletal muscle during AE, we used C2C12 myotubes, treated with the non-selective β-AR agonist isoproterenol (ISO) in the presence of the specific β2-AR antagonist ICI or during protein kinase A (PKA) and Gαi protein blockade. Our in vitro data show that β-AR activation significantly increases mitochondrial respiration in myotubes, and this response was dependent on β2-AR activation through a Gαs-PKA signaling cascade. In conclusion, we provide evidence for AE-induced β2-AR activation as a major mechanism leading to alterations in mitochondria function and morphology/dynamics. β2-AR signaling is thus a key-signaling pathway that contributes to skeletal muscle plasticity in response to exercise.
Collapse
Affiliation(s)
- Vanessa Azevedo Voltarelli
- Department of Biodynamics of the Human Body Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, SP, Brazil; (V.A.V.); (L.G.F.); (P.R.J.)
| | - Michael Coronado
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA 94304, USA; (M.C.); (G.F.)
| | - Larissa Gonçalves Fernandes
- Department of Biodynamics of the Human Body Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, SP, Brazil; (V.A.V.); (L.G.F.); (P.R.J.)
| | - Juliane Cruz Campos
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-030, SP, Brazil; (J.C.C.); (J.C.B.F.)
| | - Paulo Roberto Jannig
- Department of Biodynamics of the Human Body Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, SP, Brazil; (V.A.V.); (L.G.F.); (P.R.J.)
| | - Julio Cesar Batista Ferreira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-030, SP, Brazil; (J.C.C.); (J.C.B.F.)
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Giovanni Fajardo
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA 94304, USA; (M.C.); (G.F.)
| | - Patricia Chakur Brum
- Department of Biodynamics of the Human Body Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, SP, Brazil; (V.A.V.); (L.G.F.); (P.R.J.)
- Correspondence: or (P.C.B.); (D.B.); Tel.: +55-11-30913136 (P.C.B.); Fax: +55-11-38135921 (P.C.B.)
| | - Daniel Bernstein
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA 94304, USA; (M.C.); (G.F.)
- Correspondence: or (P.C.B.); (D.B.); Tel.: +55-11-30913136 (P.C.B.); Fax: +55-11-38135921 (P.C.B.)
| |
Collapse
|
9
|
Rodrigues ACZ, Messi ML, Wang ZM, Abba MC, Pereyra A, Birbrair A, Zhang T, O’Meara M, Kwan P, Lopez EIS, Willis MS, Mintz A, Files DC, Furdui C, Oppenheim RW, Delbono O. The sympathetic nervous system regulates skeletal muscle motor innervation and acetylcholine receptor stability. Acta Physiol (Oxf) 2019; 225:e13195. [PMID: 30269419 PMCID: PMC7224611 DOI: 10.1111/apha.13195] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
AIM Symptoms of autonomic failure are frequently the presentation of advanced age and neurodegenerative diseases that impair adaptation to common physiologic stressors. The aim of this work was to examine the interaction between the sympathetic and motor nervous system, the involvement of the sympathetic nervous system (SNS) in neuromuscular junction (NMJ) presynaptic motor function, the stability of postsynaptic molecular organization, and the skeletal muscle composition and function. METHODS Since muscle weakness is a symptom of diseases characterized by autonomic dysfunction, we studied the impact of regional sympathetic ablation on muscle motor innervation by using transcriptome analysis, retrograde tracing of the sympathetic outflow to the skeletal muscle, confocal and electron microscopy, NMJ transmission by electrophysiological methods, protein analysis, and state of the art microsurgical techniques, in C57BL6, MuRF1KO and Thy-1 mice. RESULTS We found that the SNS regulates motor nerve synaptic vesicle release, skeletal muscle transcriptome, muscle force generated by motor nerve activity, axonal neurofilament phosphorylation, myelin thickness, and myofibre subtype composition and CSA. The SNS also modulates the levels of postsynaptic membrane acetylcholine receptor by regulating the Gαi2 -Hdac4-Myogenin-MuRF1pathway, which is prevented by the overexpression of the guanine nucleotide-binding protein Gαi2 (Q205L), a constitutively active mutant G protein subunit. CONCLUSION The SNS regulates NMJ transmission, maintains optimal Gαi2 expression, and prevents any increase in Hdac4, myogenin, MuRF1, and miR-206. SNS ablation leads to upregulation of MuRF1, muscle atrophy, and downregulation of postsynaptic AChR. Our findings are relevant to clinical conditions characterized by progressive decline of sympathetic innervation, such as neurodegenerative diseases and aging.
Collapse
Affiliation(s)
- Anna C. Z. Rodrigues
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria L. Messi
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Martin C. Abba
- Basic and Applied Immunological Research Center (CINIBA), School of Medicine, National University of La Plata, Buenos Aires, Argentina
| | - Andrea Pereyra
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Alexander Birbrair
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Meaghan O’Meara
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ping Kwan
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Elsa I. S. Lopez
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Monte S. Willis
- Department of Pathology, McAllister Heart Institute, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - D. Clark Files
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Pulmonary, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Cristina Furdui
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ronald W. Oppenheim
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
10
|
Ravi V, Jain A, Ahamed F, Fathma N, Desingu PA, Sundaresan NR. Systematic evaluation of the adaptability of the non-radioactive SUnSET assay to measure cardiac protein synthesis. Sci Rep 2018; 8:4587. [PMID: 29545554 PMCID: PMC5854694 DOI: 10.1038/s41598-018-22903-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 03/01/2018] [Indexed: 11/09/2022] Open
Abstract
Heart is a dynamic organ that undergoes remodeling in response to both physiological and pathological stimuli. One of the fundamental cellular processes that facilitates changes in the size and shape of this muscular organ is the protein synthesis. Traditionally changes in cardiac protein synthesis levels were measured by radiolabeled tracers. However, these methods are often cumbersome and suffer from radioactive risk. Recently a nonradioactive method for detecting protein synthesis under in vitro conditions called the Surface Sensing of Translation (SUnSET) was described in cell lines of mouse dendrites and T cells. In this work, we provide multiple lines of evidence that the SUnSET assay can be applied to reliably detect changes in protein synthesis both in isolated neonatal primary cardiomyocytes and heart. We successfully tracked the changes in protein synthesis by western blotting as well as immunohistochemical variants of the SUnSET assay. Applying the SUnSET assay, we measured the cardiac protein synthesis during the different ages of mice. Further, we successfully tracked the increase in cardiac protein synthesis during different stages of a well-established model for pathological hypertrophy. Overall, we propose SUnSET assay as a simple, reliable and robust method to measure protein synthesis in the cardiac milieu.
Collapse
Affiliation(s)
- Venkatraman Ravi
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Aditi Jain
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Faiz Ahamed
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Nowrin Fathma
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Perumal Arumugam Desingu
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Nagalingam R Sundaresan
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India. .,Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
11
|
Miller BF, Hamilton KL, Majeed ZR, Abshire SM, Confides AL, Hayek AM, Hunt ER, Shipman P, Peelor FF, Butterfield TA, Dupont‐Versteegden EE. Enhanced skeletal muscle regrowth and remodelling in massaged and contralateral non-massaged hindlimb. J Physiol 2018; 596:83-103. [PMID: 29090454 PMCID: PMC5746529 DOI: 10.1113/jp275089] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/16/2017] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Muscle fibre cross sectional area is enhanced with massage in the form of cyclic compressive loading during regrowth after atrophy. Massage enhances protein synthesis of the myofibrillar and cytosolic, but not the mitochondrial fraction, in muscle during regrowth. Focal adhesion kinase activation and satellite cell number are elevated in muscles undergoing massage during regrowth. Muscle fibre cross sectional area and protein synthesis of the myofibrillar fraction, but not DNA synthesis, are elevated in muscle of the contralateral non-massaged limb. Massage in the form of cyclic compressive loading is a potential anabolic intervention during muscle regrowth after atrophy. ABSTRACT Massage, in the form of cyclic compressive loading (CCL), is associated with multiple health benefits, but its potential anabolic effect on atrophied muscle has not been investigated. We hypothesized that the mechanical activity associated with CCL induces an anabolic effect in skeletal muscle undergoing regrowth after a period of atrophy. Fischer-Brown Norway rats at 10 months of age were hindlimb unloaded for a period of 2 weeks. The rats were then allowed reambulation with CCL applied at a 4.5 N load at 0.5 Hz frequency for 30 min every other day for four bouts during a regrowth period of 8 days. Muscle fibre cross sectional area was enhanced by 18% with massage during regrowth compared to reloading alone, and this was accompanied by elevated myofibrillar and cytosolic protein as well as DNA synthesis. Focal adhesion kinase phosphorylation indicated that CCL increased mechanical stimulation, while a higher number of Pax7+ cells likely explains the elevated DNA synthesis. Surprisingly, the contralateral non-massaged limb exhibited a comparable 17% higher muscle fibre size compared to reloading alone, and myofibrillar protein synthesis, but not DNA synthesis, was also elevated. We conclude that massage in the form of CCL induces an anabolic response in muscles regrowing after an atrophy-inducing event. We suggest that massage can be used as an intervention to aid in the regrowth of muscle lost during immobilization.
Collapse
Affiliation(s)
- Benjamin F. Miller
- Health and Exercise ScienceColorado State UniversityFort CollinsCO80523‐1582USA
| | - Karyn L. Hamilton
- Health and Exercise ScienceColorado State UniversityFort CollinsCO80523‐1582USA
| | - Zana R. Majeed
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Sarah M. Abshire
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Amy L. Confides
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Amanda M. Hayek
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Emily R. Hunt
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Patrick Shipman
- Department of MathematicsColorado State UniversityFort CollinsCO80523‐1582USA
| | - Frederick F. Peelor
- Health and Exercise ScienceColorado State UniversityFort CollinsCO80523‐1582USA
| | - Timothy A. Butterfield
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Esther E. Dupont‐Versteegden
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKY40536‐0200USA
| |
Collapse
|
12
|
Manfredi LH, Lustrino D, Machado J, Silveira WA, Zanon NM, Navegantes LC, Kettelhut IC. Adrenodemedullation activates the Ca2+-dependent proteolysis in soleus muscles from rats exposed to cold. J Appl Physiol (1985) 2017; 122:317-326. [DOI: 10.1152/japplphysiol.00198.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 09/20/2016] [Accepted: 10/04/2016] [Indexed: 01/19/2023] Open
Abstract
Previous studies have shown that catecholamines in vivo and in vitro inhibit the activity of Ca2+-dependent proteolysis in skeletal muscles under basal conditions. In the present study we sought to investigate the role of catecholamines in regulating the Ca2+-dependent proteolysis in soleus and extensor digitorum longus (EDL) muscles from rats acutely exposed to cold. Overall proteolysis, the activity of proteolytic systems, protein levels and gene expression of different components of the calpain system were investigated in rats submitted to adrenodemedullation (ADMX) and exposed to cold for 24 h. ADMX drastically reduced plasma epinephrine and promoted an additional increase in the overall proteolysis, which was already increased by cold exposure. The rise in the rate of protein degradation in soleus muscles from adrenodemedullated cold-exposed rats was caused by the high activity of the Ca2+-dependent proteolysis, which was associated with the generation of a 145-kDa cleaved α-fodrin fragment, a typical calpain substrate, and lower protein levels and mRNA expression of calpastatin, the endogenous calpain inhibitor. Unlike that observed for soleus muscles, the cold-induced muscle proteolysis in EDL was not affected by ADMX. In isolated soleus muscle, clenbuterol, a selective β2-adrenoceptor agonist, reduced the basal Ca2+-dependent proteolysis and completely abolished the activation of this pathway by the cholinergic agonist carbachol. These data suggest that catecholamines released from the adrenal medulla inhibit cold-induced protein breakdown in soleus, and this antiproteolytic effect on the Ca2+-dependent proteolytic system is apparently mediated through expression of calpastatin, which leads to suppression of calpain activation. NEW & NOTEWORTHY Although many effects of the sympathetic nervous system on muscle physiology are known, the role of catecholamines in skeletal muscle protein metabolism has been scarcely studied. We suggest that catecholamines released from adrenal medulla may be of particular importance for restraining the activation of the Ca2+-dependent proteolysis in soleus muscles during acute cold exposure. This finding helps us to understand the adaptive changes that occur in skeletal muscle protein metabolism during cold stress.
Collapse
Affiliation(s)
- L. H. Manfredi
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Federal University of Fronteira Sul (UFFS), Chapecó, Santa Catarina, Brazil; and
| | - D. Lustrino
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - J. Machado
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - W. A. Silveira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - N. M. Zanon
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - L. C. Navegantes
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - I. C. Kettelhut
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Biochemistry/Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
13
|
dos Santos MP, Batistela E, Pereira MP, Paula-Gomes S, Zanon NM, Kettelhut IDC, Karatzaferi C, Andrade CMB, de França SA, Baviera AM, Kawashita NH. Higher insulin sensitivity in EDL muscle of rats fed a low-protein, high-carbohydrate diet inhibits the caspase-3 and ubiquitin-proteasome proteolytic systems but does not increase protein synthesis. J Nutr Biochem 2016; 34:89-98. [DOI: 10.1016/j.jnutbio.2016.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/12/2016] [Accepted: 04/23/2016] [Indexed: 12/11/2022]
|
14
|
Ijiri D, Ishitani K, Shimamoto S, Ishimaru Y, Ohtsuka A. The effects of intraperitoneal clenbuterol injection on protein degradation and myostatin expression differ between the sartorius and pectoral muscles of neonatal chicks. Gen Comp Endocrinol 2014; 206:111-7. [PMID: 25117456 DOI: 10.1016/j.ygcen.2014.07.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/25/2014] [Accepted: 07/26/2014] [Indexed: 11/29/2022]
Abstract
The purpose of this study was to investigate the effects of injection of the β2-adrenergic receptor agonist clenbuterol on the skeletal muscles of neonatal chicks (Gallus gallus domesticus). One-day-old chicks were randomly divided into four groups and given a single intraperitoneal injection of clenbuterol (0.01, 0.1, or 1mg/kg) or phosphate-buffered saline. Twenty-four hours after the injection, the sartorius muscles (which consist of both slow- and fast-twitch fibers) of chicks that received 0.01 or 0.1mg/kg clenbuterol were significantly heavier than those of controls, while there were no between-group differences in the weight of the pectoral muscles, which consist of only fast-twitch fibers. Muscle free N(t)-methylhistidine, regarded as an index of myofibrillar proteolysis, was decreased in the sartorius muscle of the clenbuterol-injected chicks, while it was not affected in the pectoral muscles. In the sartorius muscle of the clenbuterol-injected chicks, myostatin and atrogin-1/MAFbx mRNA expressions were decreased, while insulin-like growth factor-I was unaffected. These observations suggested, in 1-day-old chicks, clenbuterol might increase mass of the sartorius muscle by decreasing myostatin gene expression and protein degradation.
Collapse
Affiliation(s)
- Daichi Ijiri
- Department of Biochemical Science and Technology, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.
| | - Kanae Ishitani
- Department of Biochemical Science and Technology, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Saki Shimamoto
- Department of Biochemical Science and Technology, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Yoshitaka Ishimaru
- Department of Biochemical Science and Technology, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Akira Ohtsuka
- Department of Biochemical Science and Technology, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| |
Collapse
|
15
|
Batistela E, Pereira MP, Siqueira JT, Paula-Gomes S, Zanon NM, Oliveira EB, Navegantes LCC, Kettelhut IC, Andrade CMB, Kawashita NH, Baviera AM. Decreased rate of protein synthesis, caspase-3 activity, and ubiquitin–proteasome proteolysis in soleus muscles from growing rats fed a low-protein, high-carbohydrate diet. Can J Physiol Pharmacol 2014; 92:445-54. [DOI: 10.1139/cjpp-2013-0290] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study was to investigate the changes in the rates of both protein synthesis and breakdown, and the activation of intracellular effectors that control these processes in soleus muscles from growing rats fed a low-protein, high-carbohydrate (LPHC) diet for 15 days. The mass and the protein content, as well as the rate of protein synthesis, were decreased in the soleus from LPHC-fed rats. The availability of amino acids was diminished, since the levels of various essential amino acids were decreased in the plasma of LPHC-fed rats. Overall rate of proteolysis was also decreased, explained by reductions in the mRNA levels of atrogin-1 and MuRF-1, ubiquitin conjugates, proteasome activity, and in the activity of caspase-3. Soleus muscles from LPHC-fed rats showed increased insulin sensitivity, with increased levels of insulin receptor and phosphorylation levels of AKT, which probably explains the inhibition of both the caspase-3 activity and the ubiquitin–proteasome system. The fall of muscle proteolysis seems to represent an adaptive response that contributes to spare proteins in a condition of diminished availability of dietary amino acids. Furthermore, the decreased rate of protein synthesis may be the driving factor to the lower muscle mass gain in growing rats fed the LPHC diet.
Collapse
Affiliation(s)
- Emanuele Batistela
- Department of Chemistry, Federal University of Mato Grosso, Cuiabá, Mato Grosso, Brazil
| | - Mayara Peron Pereira
- Department of Chemistry, Federal University of Mato Grosso, Cuiabá, Mato Grosso, Brazil
| | | | - Silvia Paula-Gomes
- Department of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Neusa Maria Zanon
- Department of Physiology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Eduardo Brandt Oliveira
- Department of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Isis C. Kettelhut
- Department of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Nair Honda Kawashita
- Department of Chemistry, Federal University of Mato Grosso, Cuiabá, Mato Grosso, Brazil
| | - Amanda Martins Baviera
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University, Rua Expedicionários do Brasil, 1621, CEP 14801 360, Araraquara, São Paulo, Brazil
| |
Collapse
|
16
|
Silveira WA, Gonçalves DA, Graça FA, Andrade-Lopes AL, Bergantin LB, Zanon NM, Godinho RO, Kettelhut IC, Navegantes LCC. Activating cAMP/PKA signaling in skeletal muscle suppresses the ubiquitin-proteasome-dependent proteolysis: implications for sympathetic regulation. J Appl Physiol (1985) 2014; 117:11-9. [PMID: 24833777 DOI: 10.1152/japplphysiol.01055.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Although we have recently demonstrated that plasma catecholamines induce antiproteolytic effects on skeletal muscle (Graça FA, Gonçalves DAP, Silveira WA, Lira EC, Chaves VE, Zanon NM, Garófalo MAR, Kettelhut IC, Navegantes LCC. Am J Physiol Endocrinol Metab. 305: E1483-E1494, 2013), the role of the muscle sympathetic innervation and, more specifically, norepinephrine (NE) in regulating the ubiquitin (Ub)-proteasome system (UPS) remains unknown. Based on previous findings that chemical sympathectomy acutely reduces UPS activity, we hypothesized that muscle NE depletion induces adrenergic supersensitivity in rat skeletal muscles. We report that surgical sympathetic denervation (SDEN), a condition in which only muscle NE from both hindlimbs is depleted, transiently reduced the overall proteolysis and the UPS activity (∼25%) in both soleus and extensor digitorum longus muscles. This antiproteolytic response was accompanied by increased activity of adenylyl cyclase (112%), levels of cyclic adenosine monophosphate (cAMP; 191%), and the serine phosphorylation of cAMP response element-binding protein (32%). In extensor digitorum longus from normal rats, NE (10(-4) M) in vitro increased the levels of cAMP (115%) and the serine phosphorylation of both cAMP response element-binding protein (2.7-fold) and forkhead box class O1 transcription factor. Similar effects were observed in C2C12 cells incubated with forskolin (10 μM). In parallel, NE significantly reduced the basal UPS (21%) activity and the mRNA levels of atrophy-related Ub-ligases. Similar responses were observed in isolated muscles exposed to 6-BNZ-cAMP (500 μM), a specific PKA activator. The phosphorylation levels of Akt were not altered by SDEN, NE, forskolin or 6-BNZ-cAMP. Our results demonstrate that SDEN induces muscle adrenergic supersensitivity for cAMP leading to the suppression of UPS, and that the suppressive effects of NE on UPS activity and expression of Ub-ligases can be mediated by the activation of cAMP/PKA signaling, with the inhibition of forkhead box class O1 transcription factor.
Collapse
Affiliation(s)
- W A Silveira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - D A Gonçalves
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - F A Graça
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - A L Andrade-Lopes
- Division of Cellular Pharmacology, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - L B Bergantin
- Division of Cellular Pharmacology, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - N M Zanon
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - R O Godinho
- Division of Cellular Pharmacology, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - I C Kettelhut
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Biochemistry/Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; and
| | - L C C Navegantes
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil;
| |
Collapse
|
17
|
McDonald FB, Skelly JR, O'Halloran KD. The β2 -adrenoceptor agonist terbutaline recovers rat pharyngeal dilator muscle force decline during severe hypoxia. Oral Dis 2014; 21:e121-7. [PMID: 24725067 DOI: 10.1111/odi.12247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 03/28/2014] [Accepted: 04/08/2014] [Indexed: 11/28/2022]
Abstract
RATIONALE Obstructive sleep apnoea syndrome (OSAS) is a debilitating condition characterized by recurrent occlusions of the pharyngeal airway during sleep accompanied by arterial hypoxaemia. Upper airway muscle dysfunction is implicated in the pathophysiology of OSAS. Pharmacological agents that improve muscle contractile and endurance properties may have therapeutic value. AIM We tested the hypothesis that the β(2) -adrenoceptor agonist terbutaline improves rat sternohyoid muscle performance especially during hypoxic stress. METHODS Isometric contractile and endurance properties were examined ex vivo in Krebs solution at 35°C. Muscles were incubated in tissue baths under hyperoxic (95% O(2) /5% CO(2)) conditions in the absence (control) or presence of the β(2) -adrenoceptor agonist terbutaline (1 μM). In additional experiments under hypoxic (95% N(2) /5% CO(2)) conditions, the effects of terbutaline were examined in the presence of the β-adrenoceptor antagonist propranolol (1 μM). RESULTS Hypoxia significantly impaired sternohyoid force production. Terbutaline completely recovered hypoxic depression of force, an effect that was blocked by co-application with propranolol. CONCLUSION The β(2) -adrenoceptor agonist terbutaline completely recovers hypoxic depression of upper airway muscle force. β(2) -adrenoceptor agonists warrant investigation in animal models of OSAS reporting upper airway and diaphragm muscle dysfunction.
Collapse
Affiliation(s)
- F B McDonald
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
18
|
Graça FA, Gonçalves DAP, Silveira WA, Lira EC, Chaves VE, Zanon NM, Garófalo MAR, Kettelhut IC, Navegantes LCC. Epinephrine depletion exacerbates the fasting-induced protein breakdown in fast-twitch skeletal muscles. Am J Physiol Endocrinol Metab 2013; 305:E1483-94. [PMID: 24169047 DOI: 10.1152/ajpendo.00267.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The physiological role of epinephrine in the regulation of skeletal muscle protein metabolism under fasting is unknown. We examined the effects of plasma epinephrine depletion, induced by adrenodemedullation (ADMX), on muscle protein metabolism in fed and 2-day-fasted rats. In fed rats, ADMX for 10 days reduced muscle mass, the cross-sectional area of extensor digitorum longus (EDL) muscle fibers, and the phosphorylation levels of Akt. In addition, ADMX led to a compensatory increase in muscle sympathetic activity, as estimated by the rate of norepinephrine turnover; this increase was accompanied by high rates of muscle protein synthesis. In fasted rats, ADMX exacerbated fasting-induced proteolysis in EDL but did not affect the low rates of protein synthesis. Accordingly, ADMX activated lysosomal proteolysis and further increased the activity of the ubiquitin (Ub)-proteasome system (UPS). Moreover, expression of the atrophy-related Ub ligases atrogin-1 and MuRF1 and the autophagy-related genes LC3b and GABARAPl1 were upregulated in EDL muscles from ADMX-fasted rats compared with sham-fasted rats, and ADMX reduced cAMP levels and increased fasting-induced Akt dephosphorylation. Unlike that observed for EDL muscles, soleus muscle proteolysis and Akt phosphorylation levels were not affected by ADMX. In isolated EDL, epinephrine reduced the basal UPS activity and suppressed overall proteolysis and atrogin-1 and MuRF1 induction following fasting. These data suggest that epinephrine released from the adrenal medulla inhibits fasting-induced protein breakdown in fast-twitch skeletal muscles, and these antiproteolytic effects on the UPS and lysosomal system are apparently mediated through a cAMP-Akt-dependent pathway, which suppresses ubiquitination and autophagy.
Collapse
Affiliation(s)
- Flávia A Graça
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Rudolf R, Khan MM, Lustrino D, Labeit S, Kettelhut IC, Navegantes LCC. Alterations of cAMP-dependent signaling in dystrophic skeletal muscle. Front Physiol 2013; 4:290. [PMID: 24146652 PMCID: PMC3797997 DOI: 10.3389/fphys.2013.00290] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/24/2013] [Indexed: 12/19/2022] Open
Abstract
Autonomic regulation processes in striated muscles are largely mediated by cAMP/PKA-signaling. In order to achieve specificity of signaling its spatial-temporal compartmentation plays a critical role. We discuss here how specificity of cAMP/PKA-signaling can be achieved in skeletal muscle by spatio-temporal compartmentation. While a microdomain containing PKA type I in the region of the neuromuscular junction (NMJ) is important for postsynaptic, activity-dependent stabilization of the nicotinic acetylcholine receptor (AChR), PKA type I and II microdomains in the sarcomeric part of skeletal muscle are likely to play different roles, including the regulation of muscle homeostasis. These microdomains are due to specific A-kinase anchoring proteins, like rapsyn and myospryn. Importantly, recent evidence indicates that compartmentation of the cAMP/PKA-dependent signaling pathway and pharmacological activation of cAMP production are aberrant in different skeletal muscles disorders. Thus, we discuss here their potential as targets for palliative treatment of certain forms of dystrophy and myasthenia. Under physiological conditions, the neuropeptide, α-calcitonin-related peptide, as well as catecholamines are the most-mentioned natural triggers for activating cAMP/PKA signaling in skeletal muscle. While the precise domains and functions of these first messengers are still under investigation, agonists of β2-adrenoceptors clearly exhibit anabolic activity under normal conditions and reduce protein degradation during atrophic periods. Past and recent studies suggest direct sympathetic innervation of skeletal muscle fibers. In summary, the organization and roles of cAMP-dependent signaling in skeletal muscle are increasingly understood, revealing crucial functions in processes like nerve-muscle interaction and muscle trophicity.
Collapse
Affiliation(s)
- Rüdiger Rudolf
- Institute of Molecular and Cell Biology, University of Applied Sciences Mannheim , Mannheim, Germany ; Institute of Toxicology and Genetics, Karlsruhe Institute of Technology , Eggenstein-Leopoldshafen, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Joassard OR, Amirouche A, Gallot YS, Desgeorges MM, Castells J, Durieux AC, Berthon P, Freyssenet DG. Regulation of Akt-mTOR, ubiquitin-proteasome and autophagy-lysosome pathways in response to formoterol administration in rat skeletal muscle. Int J Biochem Cell Biol 2013; 45:2444-55. [PMID: 23916784 DOI: 10.1016/j.biocel.2013.07.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/10/2013] [Accepted: 07/24/2013] [Indexed: 10/26/2022]
Abstract
Administration of β2-agonists triggers skeletal muscle anabolism and hypertrophy. We investigated the time course of the molecular events responsible for rat skeletal muscle hypertrophy in response to 1, 3 and 10 days of formoterol administration (i.p. 2000μg/kg/day). A marked hypertrophy of rat tibialis anterior muscle culminated at day 10. Phosphorylation of Akt, ribosomal protein S6, 4E-BP1 and ERK1/2 was increased at day 3, but returned to control level at day 10. This could lead to a transient increase in protein translation and could explain previous studies that reported increase in protein synthesis following β2-agonist administration. Formoterol administration was also associated with a significant reduction in MAFbx/atrogin-1 mRNA level (day 3), suggesting that formoterol can also affect protein degradation of MAFbx/atrogin1 targeted substrates, including MyoD and eukaryotic initiation factor-3f (eIF3-f). Surprisingly, mRNA level of autophagy-related genes, light chain 3 beta (LC3b) and gamma-aminobutyric acid receptor-associated protein-like 1 (Gabarapl1), as well as lysosomal hydrolases, cathepsin B and cathepsin L, was significantly and transiently increased after 1 and/or 3 days, suggesting that autophagosome formation would be increased in response to formoterol administration. However, this has to be relativized since the mRNA level of Unc-51-like kinase1 (Ulk1), BCL2/adenovirus E1B interacting protein3 (Bnip3), and transcription factor EB (TFEB), as well as the protein content of Ulk1, Atg13, Atg5-Atg12 complex and p62/Sqstm1 remained unchanged or was even decreased in response to formoterol administration. These results demonstrate that the effects of formoterol are mediated, in part, through the activation of Akt-mTOR pathway and that other signaling pathways become more important in the regulation of skeletal muscle mass with chronic administration of β2-agonists.
Collapse
Affiliation(s)
- Olivier Roger Joassard
- Laboratoire de Physiologie de l'Exercice, Université de Lyon, F-42023 Saint-Etienne, France
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Joassard OR, Durieux AC, Freyssenet DG. β2-Adrenergic agonists and the treatment of skeletal muscle wasting disorders. Int J Biochem Cell Biol 2013; 45:2309-21. [PMID: 23845739 DOI: 10.1016/j.biocel.2013.06.025] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/14/2013] [Accepted: 06/26/2013] [Indexed: 02/04/2023]
Abstract
β2-Agonists are traditionally used for the treatment of bronchospasm associated with asthma and the treatment of symptomatic patients with COPD. However, β2-agonists are also powerful anabolic agents that trigger skeletal muscle hypertrophy. Investigating the effects of β2-agonists in skeletal muscle over the past 30 years in different animal models has led to the identification of potential therapeutic applications in several muscle wasting disorders, including neuromuscular diseases, cancer cachexia, sepsis or thermal injury. In these conditions, numerous studies indicate that β2-agonists can attenuate and/or reverse the decrease in skeletal muscle mass and associated weakness in animal models of muscle wasting but also in human patients. The purpose of this review is to present the biological and clinical significance of β2-agonists for the treatment of skeletal muscle wasting. After the description of the molecular mechanisms involved in the hypertrophy and anti-atrophy effect of β2-agonists, we will review the anti-atrophy effects of β2-agonist administration in several animal models and human pathologies associated with or leading to skeletal muscle wasting. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- Olivier R Joassard
- Laboratoire de Physiologie de l'Exercice, Université de Lyon, F-42023 Saint-Etienne, France
| | | | | |
Collapse
|
22
|
D'Hulst G, Jamart C, Van Thienen R, Hespel P, Francaux M, Deldicque L. Effect of acute environmental hypoxia on protein metabolism in human skeletal muscle. Acta Physiol (Oxf) 2013; 208:251-64. [PMID: 23418947 DOI: 10.1111/apha.12086] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 12/30/2012] [Accepted: 02/14/2013] [Indexed: 01/06/2023]
Abstract
UNLABELLED Hypoxia-induced muscle wasting has been observed in several environmental and pathological conditions. However, the molecular mechanisms behind this loss of muscle mass are far from being completely elucidated, certainly in vivo. When studying the regulation of muscle mass by environmental hypoxia, many confounding factors have to be taken into account, such as decreased protein ingestion, sleep deprivation or reduced physical activity, which make difficult to know whether hypoxia per se causes a reduction in muscle mass. AIM We hypothesized that acute exposure to normobaric hypoxia (11% O2 ) would repress the activation of the mTOR pathway usually observed after a meal and would activate the proteolytic pathways in skeletal muscle. METHODS Fifteen subjects were exposed passively for 4 h to normoxic and hypoxic conditions in a random order after consumption of a light breakfast. A muscle biopsy and a blood sample were taken before, after 1 and 4 h of exposure. RESULTS After 4 h, plasma insulin concentration and the phosphorylation state of PKB and S6K1 in skeletal muscle were higher in hypoxia than in normoxia (P < 0.05). At the same time, Redd1 mRNA level was upregulated (P < 0.05), whilst MAFbx mRNA decreased (P < 0.05) in hypoxia compared with normoxia. Proteasome, cathepsin L and calpain activities were not altered by environmental hypoxia. CONCLUSION Contrary to our hypothesis and despite an increase in the mRNA level of Redd1, an inhibitor of the mTORC1 pathway, short-term acute environmental hypoxia induced a higher response of PKB and S6K1 to a meal, which may be due to increased plasma insulin concentration.
Collapse
Affiliation(s)
- G. D'Hulst
- Department of Kinesiology; Exercise Physiology Research Group; FaBeR; KU Leuven; Leuven; Belgium
| | - C. Jamart
- Institute of Neuroscience; Université catholique de Louvain; Louvain-la-Neuve; Belgium
| | - R. Van Thienen
- Department of Kinesiology; Exercise Physiology Research Group; FaBeR; KU Leuven; Leuven; Belgium
| | - P. Hespel
- Department of Kinesiology; Exercise Physiology Research Group; FaBeR; KU Leuven; Leuven; Belgium
| | - M. Francaux
- Institute of Neuroscience; Université catholique de Louvain; Louvain-la-Neuve; Belgium
| | - L. Deldicque
- Department of Kinesiology; Exercise Physiology Research Group; FaBeR; KU Leuven; Leuven; Belgium
| |
Collapse
|
23
|
Lima MRM, Pires W, Fonseca IAT, Fonseca CG, Martinelli PM, Wanner SP, Lima NRV. Chronic sympathectomy of the caudal artery delays cutaneous heat loss during passive heating. Neurosci Lett 2013; 537:11-6. [DOI: 10.1016/j.neulet.2013.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 12/18/2012] [Accepted: 01/07/2013] [Indexed: 11/28/2022]
|
24
|
Loenneke J, Abe T, Wilson J, Thiebaud R, Fahs C, Rossow L, Bemben M. Blood flow restriction: An evidence based progressive model (Review). ACTA ACUST UNITED AC 2012; 99:235-50. [DOI: 10.1556/aphysiol.99.2012.3.1] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
25
|
Gonçalves DAP, Silveira WA, Lira EC, Graça FA, Paula-Gomes S, Zanon NM, Kettelhut IC, Navegantes LCC. Clenbuterol suppresses proteasomal and lysosomal proteolysis and atrophy-related genes in denervated rat soleus muscles independently of Akt. Am J Physiol Endocrinol Metab 2012; 302:E123-33. [PMID: 21952035 DOI: 10.1152/ajpendo.00188.2011] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although it is well known that administration of the selective β(2)-adrenergic agonist clenbuterol (CB) protects muscle following denervation (DEN), the underlying molecular mechanism remains unclear. We report that in vivo treatment with CB (3 mg/kg sc) for 3 days induces antiproteolytic effects in normal and denervated rat soleus muscle via distinct mechanisms. In normal soleus muscle, CB treatment stimulates protein synthesis, inhibits Ca(2+)-dependent proteolysis, and increases the levels of calpastatin protein. On the other hand, the administration of CB to DEN rats ameliorates the loss of muscle mass, enhances the rate of protein synthesis, attenuates hyperactivation of proteasomal and lysosomal proteolysis, and suppresses the transcription of the lysosomal protease cathepsin L and of atrogin-1/MAFbx and MuRF1, two ubiquitin (Ub) ligases involved in muscle atrophy. These effects were not associated with alterations in either IGF-I content or Akt phosphorylation levels. In isolated muscles, CB (10(-6) M) treatment significantly attenuated DEN-induced overall proteolysis and upregulation in the mRNA levels of the Ub ligases. Similar responses were observed in denervated muscles exposed to 6-BNZ-cAMP (500 μM), a PKA activator. The in vitro addition of triciribine (10 μM), a selective Akt inhibitor, did not block the inhibitory effects of CB on proteolysis and Ub ligase mRNA levels. These data indicate that short-term treatment with CB mitigates DEN-induced atrophy of the soleus muscle through the stimulation of protein synthesis, downregulation of cathepsin L and Ub ligases, and consequent inhibition of lysosomal and proteasomal activities and that these effects are independent of Akt and possibly mediated by the cAMP/PKA signaling pathway.
Collapse
Affiliation(s)
- Dawit A P Gonçalves
- Dept. of Physiology, School of Medicine, Ribeirão Preto University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Sato S, Nomura S, Kawano F, Tanihata J, Tachiyashiki K, Imaizumi K. Adaptive effects of the beta2-agonist clenbuterol on expression of beta2-adrenoceptor mRNA in rat fast-twitch fiber-rich muscles. J Physiol Sci 2010; 60:119-27. [PMID: 20033361 PMCID: PMC10716947 DOI: 10.1007/s12576-009-0075-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 11/23/2009] [Indexed: 10/20/2022]
Abstract
Administration of the beta(2)-agonist clenbuterol has been shown to reduce the expression of beta(2)-adrenoceptor (AR) mRNA in fast-twitch fiber-rich (extensor digitorum longus, EDL) muscle without changing that in slow-twitch fiber-rich (soleus, SOL) muscle in rats. However, the regulatory mechanism for muscle fiber type-dependent down-regulation of the expression of beta(2)-AR mRNA induced by clenbuterol is still unclear. Therefore, mRNA expression of transcriptional and post-transcriptional regulatory factors for beta(2)-AR mRNA levels in fast-twitch fiber-rich (EDL and plantaris, PLA) and slow-twitch fiber-rich (SOL) muscles in clenbuterol-administered (1.0 mg/kg body weight/day for 10 days, subcutaneous) rats was studied by real-time reverse transcription-polymerase chain reaction. Administration of clenbuterol significantly reduced expression of beta(2)-AR mRNA in EDL and PLA muscles without changing that in SOL muscle. Administration of clenbuterol also significantly reduced the mRNA expression of transcriptional regulatory factor (glucocorticoid receptor) and mRNA stabilizing factor (Hu antigen R) in EDL and PLA muscles without changing those in SOL muscle. These results suggest that muscle fiber type-dependent effects of clenbuterol on expression of beta(2)-AR mRNA are closely related to the down-regulation of mRNA expression of transcriptional and post-transcriptional regulatory factors for beta(2)-AR mRNA levels.
Collapse
MESH Headings
- Adaptation, Physiological/drug effects
- Adaptation, Physiological/physiology
- Adrenergic beta-Agonists/pharmacology
- Animals
- Cells, Cultured
- Clenbuterol/pharmacology
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Male
- Muscle Fibers, Fast-Twitch/drug effects
- Muscle Fibers, Fast-Twitch/physiology
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/metabolism
Collapse
Affiliation(s)
- Shogo Sato
- Laboratory of Physiological Sciences, Faculty of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192 Japan
| | - Sachiko Nomura
- Graduate School of Medicine, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka 560-0043 Japan
| | - Fuuun Kawano
- Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902 Japan
| | - Jun Tanihata
- Laboratory of Rehabilitation Biomedical Sciences, Faculty of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192 Japan
| | - Kaoru Tachiyashiki
- Department of Living and Health Sciences, Graduate School of Joetsu University of Education, 1 Yamayashiki, Joetsu, Niigata 943-8512 Japan
| | - Kazuhiko Imaizumi
- Laboratory of Physiological Sciences, Faculty of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192 Japan
| |
Collapse
|
27
|
Baviera AM, Zanon NM, Navegantes LCC, Kettelhut IC. Involvement of cAMP/Epac/PI3K-dependent pathway in the antiproteolytic effect of epinephrine on rat skeletal muscle. Mol Cell Endocrinol 2010; 315:104-12. [PMID: 19804812 DOI: 10.1016/j.mce.2009.09.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 09/14/2009] [Accepted: 09/28/2009] [Indexed: 02/04/2023]
Abstract
Very little is known about the signaling pathways by which catecholamines exert anabolic effects on muscle protein metabolism, stimulating protein synthesis and suppressing proteolysis. The present work tested the hypothesis that epinephrine-induced inhibition of muscle proteolysis is mediated through the cAMP/Epac/PI3K-dependent pathway with the involvement of AKT and Foxo. The incubation of extensor digitorum longus (EDL) muscles from rats with epinephrine and/or insulin increased the phosphorylation of AKT and its downstream target Foxo3a, a well-known effect that prevents Foxo translocation to the nucleus and the activation of proteolysis. Similar effects on AKT/Foxo signaling were observed in muscles incubated with DBcAMP (cAMP analog). The stimulatory effect of epinephrine on AKT phosphorylation was completely blocked by wortmannin (selective PI3K inhibitor), suggesting that the epinephrine-induced activation of AKT is mediated through PI3K. As for epinephrine and DBcAMP, the incubation of muscles with 8CPT-2Me-cAMP (selective Epac agonist) reduced rates of proteolysis and increased phosphorylation levels of AKT and Foxo3a. The specific PKA agonist (N6BZ-cAMP) inhibited proteolysis and abolished the epinephrine-induced AKT and Foxo3a phosphorylation. On the other hand, inhibition of PKA by H89 further increased the phosphorylation levels of AKT and Foxo3a induced by epinephrine, DBcAMP or 8CPT-2Me-cAMP. These findings suggest that the antiproteolytic effect of the epinephrine on isolated skeletal muscle may occur through a cAMP/Epac/PI3K-dependent pathway, which leads to the phosphorylation of AKT and Foxo3a. The parallel activation of PKA-dependent pathway also inhibits proteolysis and seems to limit the stimulatory effect of cAMP on AKT/Foxo3a signaling.
Collapse
Affiliation(s)
- Amanda Martins Baviera
- Department of Chemistry, Federal University of Mato Grosso, 78060-900 Cuiabá, MT, Brazil
| | | | | | | |
Collapse
|
28
|
Gonçalves DAP, Lira EC, Baviera AM, Cao P, Zanon NM, Arany Z, Bedard N, Tanksale P, Wing SS, Lecker SH, Kettelhut IC, Navegantes LCC. Mechanisms involved in 3',5'-cyclic adenosine monophosphate-mediated inhibition of the ubiquitin-proteasome system in skeletal muscle. Endocrinology 2009; 150:5395-404. [PMID: 19837877 DOI: 10.1210/en.2009-0428] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although it is well known that catecholamines inhibit skeletal muscle protein degradation, the molecular underlying mechanism remains unclear. This study was undertaken to investigate the role of beta(2)-adrenoceptors (AR) and cAMP in regulating the ubiquitin-proteasome system (UPS) in skeletal muscle. We report that increased levels of cAMP in isolated muscles, promoted by the cAMP phosphodiesterase inhibitor isobutylmethylxanthine was accompanied by decreased activity of the UPS, levels of ubiquitin-protein conjugates, and expression of atrogin-1, a key ubiquitin-protein ligase involved in muscle atrophy. In cultured myotubes, atrogin-1 induction after dexamethasone treatment was completely prevented by isobutylmethylxanthine. Furthermore, administration of clenbuterol, a selective beta(2)-agonist, to mice increased muscle cAMP levels and suppressed the fasting-induced expression of atrogin-1 and MuRF-1, atrogin-1 mRNA being much more responsive to clenbuterol. Moreover, clenbuterol increased the phosphorylation of muscle Akt and Foxo3a in fasted rats. Similar responses were observed in muscles exposed to dibutyryl-cAMP. The stimulatory effect of clenbuterol on cAMP and Akt was abolished in muscles from beta(2)-AR knockout mice. The suppressive effect of beta(2)-agonist on atrogin-1 was not mediated by PGC-1alpha (peroxisome proliferator-activated receptor-gamma coactivator 1alpha known to be induced by beta(2)-agonists and previously shown to inhibit atrogin-1 expression), because food-deprived PGC-1alpha knockout mice were still sensitive to clenbuterol. These findings suggest that the cAMP increase induced by stimulation of beta(2)-AR in skeletal muscles from fasted mice is possibly the mechanism by which catecholamines suppress atrogin-1 and the UPS, this effect being mediated via phosphorylation of Akt and thus inactivation of Foxo3.
Collapse
MESH Headings
- 1-Methyl-3-isobutylxanthine/pharmacology
- Adrenergic beta-2 Receptor Agonists
- Animals
- Blotting, Western
- Cell Line
- Clenbuterol/pharmacology
- Cyclic AMP/metabolism
- Dexamethasone/pharmacology
- Forkhead Box Protein O3
- Forkhead Transcription Factors/metabolism
- In Vitro Techniques
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
- Phosphodiesterase Inhibitors/pharmacology
- Phosphorylation/drug effects
- Proteasome Endopeptidase Complex/genetics
- Proteasome Endopeptidase Complex/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Wistar
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- SKP Cullin F-Box Protein Ligases/genetics
- SKP Cullin F-Box Protein Ligases/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors
- Tripartite Motif Proteins
- Ubiquitin/genetics
- Ubiquitin/metabolism
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
- Dawit A P Gonçalves
- Departments of Physiology and Biochemistry & Immunology, School of Medicine, University of São Paulo, 14049-900 Ribeirão Preto, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Harris RBS, Kelso EW, Flatt WP, Grill HJ, Bartness TJ. Testosterone replacement does not normalize carcass composition in chronically decerebrate male rats. Am J Physiol Regul Integr Comp Physiol 2009; 296:R1687-94. [PMID: 19369588 DOI: 10.1152/ajpregu.00019.2009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronically decerebrate (CD) rats, in which the forebrain and its descending projections are completely neurally isolated from hindbrain and rostral projections, gain substantial amounts of body fat, lose lean tissue, and have low circulating testosterone concentrations. We tested whether testosterone replacement would normalize body composition of male CD rats. Five groups of rats were used: CD placebo, CD testosterone, control placebo, castrate placebo, and castrate testosterone. Testosterone replacement was initiated at the first stage of CD surgery in both CDs and castrate controls. The second stage of CD surgery occurred 8 days later, and the study ended 15 days later. Testosterone implants produced 10-fold normal circulating concentrations. Food intake was fixed for all rats by tube feeding. CD rats had substantially more body fat and less lean tissue than neurally intact rats. Testosterone replacement did not affect adiposity of CD rats but did increase carcass water content. Energy expenditure of CD rats was significantly lower than that of control placebo and castrated rats. Testosterone lowered respiratory equivalency ratio and ameliorated a fall in energy expenditure late in the intermeal interval in CD rats. Castration increased, and testosterone decreased luteinizing hormone (LH) and follicle stimulating hormone (FSH) in neurally intact controls. LH was undetectable, and FSH was equivalent to neurally intact controls in CD rats, and neither was affected by testosterone. Collectively, low testosterone did not explain obesity or decreased lean body mass of CD rats, although CD rats exhibited abnormal levels of circulating reproductive hormones and disrupted testosterone negative feedback.
Collapse
Affiliation(s)
- Ruth B S Harris
- Department of Biology, Georgia State University, Atlanta, GA 30302-4010, USA
| | | | | | | | | |
Collapse
|
30
|
Burniston JG, McLean L, Beynon RJ, Goldspink DF. Anabolic effects of a non-myotoxic dose of the beta2-adrenergic receptor agonist clenbuterol on rat plantaris muscle. Muscle Nerve 2007; 35:217-23. [PMID: 17058275 PMCID: PMC1852641 DOI: 10.1002/mus.20684] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Previous investigations of the effects of clenbuterol have used suprapharmacological doses that induce myocyte death, alter muscle phenotype, and do not approximate the proposed therapeutic dose for humans. Recently, we reported that smaller doses of clenbuterol induce muscle growth without causing myocyte death. In the present study we used histochemical and proteomic techniques to investigate the molecular effects of this dose. Male Wistar rats (n = 6, per group) were infused with saline or 10 microg/kg/day clenbuterol via subcutaneously implanted osmotic pumps. After 14 days the animals' plantaris muscles were isolated for histochemical and proteomic analyses. Clenbuterol induced significant muscle growth with concomitant protein accretion and preferential hypertrophy of fast oxidative glycolytic fibers. Clenbuterol reduced the optical density of mitochondrial staining in fast fibers by 20% and the glycogen content of the muscle by 30%. Differential analysis of two-dimensional gels showed that heat shock protein 72 and beta-enolase increased, whereas aldolase A, phosphogylcerate mutase, and adenylate kinase decreased. Only heat shock protein 72 has previously been investigated in clenbuterol-treated muscles. The clenbuterol-induced increase in muscle growth was concomitant with qualitative changes in the muscle's proteome that need to be considered when proposing therapeutic uses for this agent.
Collapse
Affiliation(s)
- Jatin G Burniston
- Muscle Physiology and Proteomics Laboratory, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Webster Street, Liverpool L3 2ET, UK.
| | | | | | | |
Collapse
|
31
|
Baviera AM, Zanon NM, Carvalho Navegantes LC, Migliorini RH, do Carmo Kettelhut I. Pentoxifylline inhibits Ca2+-dependent and ATP proteasome-dependent proteolysis in skeletal muscle from acutely diabetic rats. Am J Physiol Endocrinol Metab 2007; 292:E702-8. [PMID: 17077345 DOI: 10.1152/ajpendo.00147.2006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies from this laboratory have shown that catecholamines exert an inhibitory effect on muscle protein degradation through a pathway involving the cAMP cascade. The present work investigated the systemic effect of pentoxifylline (PTX; cAMP-phosphodiesterase inhibitor) treatment on the rate of overall proteolysis, the activity of proteolytic systems, and the process of protein synthesis in extensor digitorum longus muscles from normal and acutely diabetic rats. The direct in vitro effect of this drug on the rates of muscle protein degradation was also investigated. Muscles from diabetic rats treated with PTX showed an increase (22%) in the cAMP content and reduction in total rates of protein breakdown and in activity of Ca2+-dependent (47%) and ATP proteasome-dependent (23%) proteolytic pathways. The high content of m-calpain observed in muscles from diabetic rats was abolished by PTX treatment. The addition of PTX (10(-3) M) to the incubation medium increased the cAMP content in muscles from normal (22%) and diabetic (51%) rats and induced a reduction in the rates of overall proteolysis that was accompanied by decreased activity of the Ca2+-dependent and ATP proteasome-dependent proteolytic systems, in both groups. The in vitro addition of H-89, an inhibitor of protein kinase A (PKA), completely blocked the effect of PTX on the reduction of proteolysis in muscles from normal and diabetic rats. The present data suggest that PTX exerts a direct inhibitory effect on protein degradative systems in muscles from acutely diabetic rats, probably involving the participation of cAMP intracellular pathways and activation of PKA, independently of tumor necrosis factor-alpha inhibition.
Collapse
Affiliation(s)
- Amanda Martins Baviera
- Department of Biochemistry and Immunology, School of Medicine, USP, 14049-900 Ribeirão Preto, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
32
|
Jones RJ, Jourd'heuil D, Salerno JC, Smith SME, Singer HA. iNOS regulation by calcium/calmodulin-dependent protein kinase II in vascular smooth muscle. Am J Physiol Heart Circ Physiol 2007; 292:H2634-42. [PMID: 17293490 DOI: 10.1152/ajpheart.01247.2006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nitric oxide synthase (NOS) expression is regulated transcriptionally in response to cytokine induction and posttranslationally by palmitoylation and trafficking into perinuclear aggresome-like structures. We investigated the effects of multifunctional calcium/calmodulin-dependent protein kinase II protein kinase (CaMKII) on inducible NOS (iNOS) trafficking in cultured rat aortic vascular smooth muscle cells (VSMCs). Immunofluorescence and confocal microscopy demonstrated colocalization of iNOS and CaMKIIdelta(2) with a perinuclear distribution and concentration in aggresome-like structures identified by colocalization with gamma-tubulin. Furthermore, CaMKIIdelta(2) coimmunoprecipitated with iNOS in a CaMKII activity-dependent manner. Addition of Ca(2+)-mobilizing stimuli expected to activate CaMKII; a purinergic agonist (UTP) or calcium ionophore (ionomycin) caused a general redistribution of iNOS from cytosolic to membrane and nuclear fractions. Similarly, adenoviral expression of a constitutively active CaMKIIdelta(2) mutant altered iNOS localization, shifting iNOS from the cytosolic fraction. Suppression of CaMKIIdelta(2) using an adenovirus expressing a short hairpin, small interfering RNA increased nuclear iNOS localization in resting cells but inhibited ionomycin-induced translocation of iNOS to the nucleus. Following addition of these chronic and acute CaMKII modulators, there were fewer aggresome-like structures containing iNOS. All of the treatments that chronically affected CaMKII activity or expression significantly inhibited iNOS-specific activity following cytokine induction. The results suggest that CaMKIIdelta(2) may be an important regulator of iNOS trafficking and activity in VSMCs.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/metabolism
- Benzylamines/pharmacology
- Calcium/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2
- Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors
- Calcium-Calmodulin-Dependent Protein Kinases/genetics
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Cell Membrane/enzymology
- Cell Nucleus/enzymology
- Cells, Cultured
- Cytokines/metabolism
- Cytoplasm/enzymology
- Enzyme Induction
- Interferon-gamma/metabolism
- Interleukin-1beta/metabolism
- Ionomycin/pharmacology
- Ionophores/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Nitric Oxide Synthase Type II/biosynthesis
- Protein Kinase Inhibitors/pharmacology
- Protein Transport
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Sulfonamides/pharmacology
- Tumor Necrosis Factor-alpha/metabolism
- Uridine Triphosphate/metabolism
Collapse
Affiliation(s)
- Rachel J Jones
- Center for Cardiovascular Sciences, Albany Medical College, 43 New Scotland Ave., Albany, NY 12208, USA
| | | | | | | | | |
Collapse
|
33
|
Kreier F, Kalsbeek A, Sauerwein HP, Fliers E, Romijn JA, Buijs RM. “Diabetes of the elderly” and type 2 diabetes in younger patients: Possible role of the biological clock. Exp Gerontol 2007; 42:22-7. [PMID: 17049784 DOI: 10.1016/j.exger.2006.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Revised: 07/02/2006] [Accepted: 07/06/2006] [Indexed: 11/16/2022]
Abstract
The increased prevalence of type 2 diabetes in the aged has been recognized for a long time. Within the last decades, a growing number of younger subjects and even children are prone to develop type 2 diabetes. In both groups, aged and young, the biological clock, located in the suprachiasmatic nucleus of the hypothalamus (SCN) is malfunctioning as evidenced by disturbed sleep cycles and altered circadian rhythms. While elderly patients have an impaired function of the SCN due to the degeneration of neurons, we propose that in younger subjects the clock loses its "feeling" for internal and external rhythms caused by the modern lifestyle. Sleeping late and less coupled with constant metabolic excess alter both internal and external environmental stimuli to the brain. In response to these alterations, the rhythm of the biological clock is disrupted which may lead to the metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- Felix Kreier
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
34
|
Burniston JG, Clark WA, Tan LB, Goldspink DF. Dose-dependent separation of the hypertrophic and myotoxic effects of the beta(2)-adrenergic receptor agonist clenbuterol in rat striated muscles. Muscle Nerve 2006; 33:655-63. [PMID: 16411205 PMCID: PMC1828609 DOI: 10.1002/mus.20504] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Muscle growth in response to large doses (milligrams per kilogram) of beta(2)-adrenergic receptor agonists has been reported consistently. However, such doses may also induce myocyte death in the heart and skeletal muscles and hence may not be safe doses for humans. We report the hypertrophic and myotoxic effects of different doses of clenbuterol. Rats were infused with clenbuterol (range, 1 microg to 1 mg.kg(-1)) for 14 days. Muscle protein content, myofiber cross-sectional area, and myocyte death were then investigated. Infusions of >or=10 microg.kg(-1).d(-1) of clenbuterol significantly (P<0.05) increased the protein content of the heart (12%-15%), soleus (12%), plantaris (18%-29%), and tibialis anterior (11%-22%) muscles, with concomitant myofiber hypertrophy. Larger doses (100 microg or 1 mg) induced significant (P<0.05) myocyte death in the soleus (peak 0.2+/-0.1% apoptosis), diaphragm (peak 0.15+/-0.1% apoptosis), and plantaris (peak 0.3+/-0.05% necrosis), and significantly increased the area fraction of collagen in the myocardium. These data show that the low dose of 10 microg.kg(-1).d(-1) can be used in rats to investigate the anabolic effects of clenbuterol in the absence of myocyte death.
Collapse
Affiliation(s)
- Jatin G Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Webster Street, Liverpool L3 2ET, UK.
| | | | | | | |
Collapse
|
35
|
Akutsu S, Shimada A, Yamane A. Transforming growth factor betas are upregulated in the rat masseter muscle hypertrophied by clenbuterol, a beta2 adrenergic agonist. Br J Pharmacol 2006; 147:412-21. [PMID: 16402040 PMCID: PMC1616986 DOI: 10.1038/sj.bjp.0706625] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. The regulatory mechanism for the hypertrophy of skeletal muscles induced by clenbuterol is unclear. The purpose of the present study was to determine the extent to which transforming growth factor betas (TGFbetas), fibroblast growth factors (FGFs), hepatocyte growth factor (HGF), and platelet-derived growth factors (PDGFs) are involved in the hypertrophy of rat masseter muscle induced by clenbuterol. 2. We measured the mRNA expression levels for TGFbetas, FGFs, HGF, and PDGFs in rat masseter muscle hypertrophied by oral administration of clenbuterol for 3 weeks and determined correlations between the weight of masseter muscle and mRNA expression levels by regression analysis. We determined immunolocalizations of TGFbetas and their receptors (TGFbetaRs). 3. The mRNA expression levels for TGFbeta1, 2, and 3, and for PDGF-B demonstrated clenbuterol-induced elevations and positive correlations with the weight of masseter muscle. In particular, TGFbeta1, 2, and 3 showed strong positive correlations (correlation coefficients >0.6). The mRNA expression levels for PDGF-A, FGF-1 and 2, and HGF showed no significant differences between the control and clenbuterol groups, and no significant correlations. TGFbeta1, 2, and 3 were principally localized in the connective tissues interspaced among myofibers, and TGFbetaRI and II were localized in the periphery and sarcoplasm of the myofibers. 4. These results suggest that paracrine actions of TGFbeta1, 2, and 3 via TGFbetaRI and II could be involved in the hypertrophy of rat masseter muscle induced by clenbuterol. This is the first study to document the involvement of TGFbetas in the hypertrophy of skeletal muscles induced by clenbuterol.
Collapse
Affiliation(s)
- Satonari Akutsu
- Katayanagi Advanced Research Laboratories, Tokyo University of Technology, Hachioji, Tokyo, Japan
| | - Akemi Shimada
- Department of Pharmacology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan
| | - Akira Yamane
- Department of Pharmacology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan
- Author for correspondence:
| |
Collapse
|
36
|
Kreier F, Kap YS, Mettenleiter TC, van Heijningen C, van der Vliet J, Kalsbeek A, Sauerwein HP, Fliers E, Romijn JA, Buijs RM. Tracing from fat tissue, liver, and pancreas: a neuroanatomical framework for the role of the brain in type 2 diabetes. Endocrinology 2006; 147:1140-7. [PMID: 16339209 DOI: 10.1210/en.2005-0667] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The hypothalamus uses hormones and the autonomic nervous system to balance energy fluxes in the body. Here we show that the autonomic nervous system has a distinct organization in different body compartments. The same neurons control intraabdominal organs (intraabdominal fat, liver, and pancreas), whereas sc adipose tissue located outside the abdominal compartment receives input from another set of autonomic neurons. This differentiation persists up to preautonomic neurons in the hypothalamus, including the biological clock, that have a distinct organization depending on the body compartment they command. Moreover, we demonstrate a neuronal feedback from adipose tissue that reaches the brainstem. We propose that this compartment-specific organization offers a neuroanatomical perspective for the regional malfunction of organs in type 2 diabetes, where increased insulin secretion by the pancreas and disturbed glucose metabolism in the liver coincide with an augmented metabolic activity of visceral compared with sc adipose tissue.
Collapse
Affiliation(s)
- Felix Kreier
- Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Navegantes LCC, Resano NMZ, Baviera AM, Migliorini RH, Kettelhut IC. CL 316,243, a selective β3-adrenergic agonist, inhibits protein breakdown in rat skeletal muscle. Pflugers Arch 2005; 451:617-24. [PMID: 16091956 DOI: 10.1007/s00424-005-1496-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2005] [Revised: 07/01/2005] [Accepted: 07/14/2005] [Indexed: 10/25/2022]
Abstract
The in vitro effect of CL 316,243 (CL), a selective beta3-adrenoceptor agonist in the rate of overall proteolysis, the activity of proteolytic systems (lysosomal, Ca2+-dependent, ATP-dependent, and ATP-independent) and in the process of protein synthesis was investigated in rat skeletal muscles. The rate of overall proteolysis in soleus muscle from rats incubated with CL (10(-4) and 10(-5) M) or epinephrine (10(-5) M) was significantly decreased. In vitro rates of maximal activity of Ca2+-dependent proteolysis in soleus muscles were decreased by about 41% in the presence of 10(-5) M CL. No change was observed in the activities of the lysosomal, ATP-dependent or ATP-independent proteolytic systems. The anti-proteolytic effect of CL or epinephrine was partially prevented by 10(-5) M SR 59230A, a selective beta3-adrenoceptor antagonist. The increase of proteolysis induced by food deprivation in soleus was abolished by in vitro addition of 10(-5) M CL. No change in proteolysis was observed in extensor digitorum longus (EDL) muscles incubated with any concentration of the beta3-adrenoceptor agonist tested. Rates of protein synthesis were not affected by 10(-4) M CL neither in soleus nor EDL. The data suggest that a beta3-adrenoceptor-mediated inhibition of Ca2+-dependent proteolysis participates of the antiproteolytic effect of catecholamines in oxidative muscles.
Collapse
Affiliation(s)
- Luiz Carlos C Navegantes
- Department of Physiology, Biochemistry and Immunology, School of Medicine, University of São Paulo, 14049-900, Ribeirão Preto, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
38
|
Manoli I, Le H, Alesci S, McFann KK, Su YA, Kino T, Chrousos GP, Blackman MR. Monoamine oxidase-A is a major target gene for glucocorticoids in human skeletal muscle cells. FASEB J 2005; 19:1359-61. [PMID: 15946989 DOI: 10.1096/fj.04-3660fje] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Skeletal myopathy is a common complication of endogenous and exogenous glucocorticoid excess, yet its pathogenetic mechanisms remain unclear. There is accumulating evidence that mitochondrial dysfunction and oxidative stress are involved in this process. To explore the glucocorticoid-induced transcriptional adaptations that may affect mitochondrial function in skeletal muscle, we studied gene expression profiles in dexamethasone-treated primary human skeletal myocytes using a cDNA microarray, which contains 501 mitochondria-related genes. We found that monoamine oxidase A (MAO-A) was the most significantly up-regulated gene. MAO-A is the primary enzyme metabolizing catecholamines and dietary amines, and its role in skeletal muscle remains largely unexplored. Dexamethasone induced dose- and time-dependent increases of MAO-A gene and protein expression, while its effects on MAO-B were minimal. Both the glucocorticoid receptor (GR) and the Sp1 transcription factor were required for dexamethasone-induced MAO-A mRNA expression, as blockade of the GR with RU 486 or ablation of Sp1 binding with mithramycin abrogated MAO-A mRNA induction. The observed dexamethasone effect was biologically functional, as this steroid significantly increased MAO-mediated hydrogen peroxide production. We suggest that MAO-A-mediated oxidative stress can lead to cell damage, representing a novel pathogenetic mechanism for glucocorticoid-induced myopathy and a potential target for therapeutic intervention.
Collapse
MESH Headings
- Adolescent
- Adult
- Cells, Cultured
- Dexamethasone/toxicity
- Dose-Response Relationship, Drug
- Gene Expression Regulation, Enzymologic/drug effects
- Humans
- Hydrogen Peroxide/metabolism
- Male
- Monoamine Oxidase/biosynthesis
- Monoamine Oxidase/genetics
- Monoamine Oxidase Inhibitors/pharmacology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/enzymology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscular Diseases/chemically induced
- Oligonucleotide Array Sequence Analysis
- Promoter Regions, Genetic
- RNA, Messenger/analysis
- Receptors, Glucocorticoid/physiology
- Sp1 Transcription Factor/physiology
- Transcriptional Activation
Collapse
Affiliation(s)
- Irini Manoli
- Endocrine Section, Laboratory of Clinical Investigation, NCCAM, NIH, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Hinkle RT, Dolan E, Cody DB, Bauer MB, Isfort RJ. Phosphodiesterase 4 inhibition reduces skeletal muscle atrophy. Muscle Nerve 2005; 32:775-81. [PMID: 16116651 DOI: 10.1002/mus.20416] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several GTP-binding protein (G-protein)-coupled receptors that signal through Galphas (GTP-binding protein alpha stimulatory) and the cyclic adenosine monophosphate (cAMP) pathway increase skeletal muscle mass. In order to further evaluate the role of the cAMP pathway in the regulation of skeletal muscle mass, we utilized inhibitors of phosphodiesterase 4 (PDE 4), the major cAMP-modifying PDE found in skeletal muscle, to modulate skeletal muscle cAMP levels. We found that PDE 4 inhibitors reduced the loss of muscle mass and force resulting from denervation and casting in rats and mice. These studies indicate that PDE 4 inhibitors may have a role in the treatment of skeletal muscle-wasting diseases.
Collapse
Affiliation(s)
- Richard T Hinkle
- Research Division, Procter & Gamble Pharmaceuticals, Health Care Research Center, 8700 Mason-Montgomery Road, Mason, Ohio 45040, USA
| | | | | | | | | |
Collapse
|