51
|
Sharples AP, Hughes DC, Deane CS, Saini A, Selman C, Stewart CE. Longevity and skeletal muscle mass: the role of IGF signalling, the sirtuins, dietary restriction and protein intake. Aging Cell 2015; 14:511-23. [PMID: 25866088 PMCID: PMC4531066 DOI: 10.1111/acel.12342] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2015] [Indexed: 12/11/2022] Open
Abstract
Advancing age is associated with a progressive loss of skeletal muscle (SkM) mass and function. Given the worldwide aging demographics, this is a major contributor to morbidity, escalating socio-economic costs and ultimately mortality. Previously, it has been established that a decrease in regenerative capacity in addition to SkM loss with age coincides with suppression of insulin/insulin-like growth factor signalling pathways. However, genetic or pharmacological modulations of these highly conserved pathways have been observed to significantly enhance life and healthspan in various species, including mammals. This therefore provides a controversial paradigm in which reduced regenerative capacity of skeletal muscle tissue with age potentially promotes longevity of the organism. This paradox will be assessed and considered in the light of the following: (i) the genetic knockout, overexpression and pharmacological models that induce lifespan extension (e.g. IRS-1/s6K KO, mTOR inhibition) versus the important role of these signalling pathways in SkM growth and adaptation; (ii) the role of the sirtuins (SIRTs) in longevity versus their emerging role in SkM regeneration and survival under catabolic stress; (iii) the role of dietary restriction and its impact on longevity versus skeletal muscle mass regulation; (iv) the crosstalk between cellular energy metabolism (AMPK/TSC2/SIRT1) and survival (FOXO) versus growth and repair of SkM (e.g. AMPK vs. mTOR); and (v) the impact of protein feeding in combination with dietary restriction will be discussed as a potential intervention to maintain SkM mass while increasing longevity and enabling healthy aging.
Collapse
Affiliation(s)
- Adam P. Sharples
- Stem Cells, Ageing & Molecular Physiology Unit; Research Institute for Sport and Exercise Sciences (RISES); Exercise Metabolism and Adaptation Research Group (EMARG); Liverpool John Moores University; Tom Reilly Building Liverpool L3 3AF UK
| | - David C. Hughes
- Stem Cells, Ageing & Molecular Physiology Unit; Research Institute for Sport and Exercise Sciences (RISES); Exercise Metabolism and Adaptation Research Group (EMARG); Liverpool John Moores University; Tom Reilly Building Liverpool L3 3AF UK
- Department of Neurobiology, Physiology and Behavior; University of California; Davis California CA 95616 USA
| | - Colleen S. Deane
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research; School of Medicine; University of Nottingham; Royal Derby Hospital; Derby DE22 3DT UK
- School of Health and Social Care; Bournemouth University; Bournemouth BH12 5BB UK
| | - Amarjit Saini
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm 171 77 Sweden
| | - Colin Selman
- Glasgow Ageing Research Network (GARNER); Institute of Biodiversity, Animal Health and Comparative Medicine; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow G12 8QQ UK
| | - Claire E. Stewart
- Stem Cells, Ageing & Molecular Physiology Unit; Research Institute for Sport and Exercise Sciences (RISES); Exercise Metabolism and Adaptation Research Group (EMARG); Liverpool John Moores University; Tom Reilly Building Liverpool L3 3AF UK
| |
Collapse
|
52
|
Ohira T, Kawano F, Ohira T, Goto K, Ohira Y. Responses of skeletal muscles to gravitational unloading and/or reloading. J Physiol Sci 2015; 65:293-310. [PMID: 25850921 PMCID: PMC10717835 DOI: 10.1007/s12576-015-0375-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/17/2015] [Indexed: 02/04/2023]
Abstract
Adaptation of morphological, metabolic, and contractile properties of skeletal muscles to inhibition of antigravity activities by exposure to a microgravity environment or by simulation models, such as chronic bedrest in humans or hindlimb suspension in rodents, has been well reported. Such physiological adaptations are generally detrimental in daily life on earth. Since the development of suitable countermeasure(s) is essential to prevent or inhibit these adaptations, effects of neural, mechanical, and metabolic factors on these properties in both humans and animals were reviewed. Special attention was paid to the roles of the motoneurons (both efferent and afferent neurograms) and electromyogram activities as the neural factors, force development, and/or length of sarcomeres as the mechanical factors and mitochondrial bioenergetics as the metabolic factors.
Collapse
Affiliation(s)
- Takashi Ohira
- Space Biomedical Research Office, Japan Aerospace Exploration Agency, Tsukuba, Ibaraki 305-8505 Japan
| | - Fuminori Kawano
- Graduate School of Medicine, Osaka University, Toyonaka, Osaka 560-0043 Japan
| | - Tomotaka Ohira
- Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi 440-8511 Japan
| | - Katsumasa Goto
- Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi, Aichi 440-8511 Japan
| | - Yoshinobu Ohira
- Graduate School of Health and Sports Science, Doshisha University, Miyakodani 1-3, Tatara, Kyotanabe, Kyoto 610-0394 Japan
| |
Collapse
|
53
|
Houston FE, Hain BA, Adams TJ, Houston KL, O'Keeffe R, Dodd SL. Heat shock protein 70 overexpression does not attenuate atrophy in botulinum neurotoxin type A-treated skeletal muscle. J Appl Physiol (1985) 2015; 119:83-92. [PMID: 25953835 DOI: 10.1152/japplphysiol.00233.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 04/29/2015] [Indexed: 11/22/2022] Open
Abstract
Botulinum neurotoxin type A (BoNT/A) is used clinically to induce therapeutic chemical denervation of spastically contracted skeletal muscles. However, BoNT/A administration can also cause atrophy. We sought to determine whether a major proteolytic pathway contributing to atrophy in multiple models of muscle wasting, the ubiquitin proteasome system (UPS), is involved in BoNT/A-induced atrophy. Three and ten days following BoNT/A injection of rat hindlimb, soleus muscle fiber cross-sectional area was reduced 25 and 65%, respectively. The transcriptional activity of NF-κB and Foxo was significantly elevated at 3 days (2- to 4-fold) and 10 days (5- to 6-fold). Muscle RING-finger protein-1 (MuRF1) activity was elevated (2-fold) after 3 days but not 10 days, while atrogin-1 activity was not elevated at any time point. BoNT/A-induced polyubiquitination occurred after 3 days (3-fold increase) but was totally absent after 10 days. Proteasome activity was elevated (1.5- to 2-fold) after 3 and 10 days. We employed the use of heat shock protein 70 (Hsp70) to inhibit NF-κB and Foxo transcriptional activity. Electrotransfer of Hsp70 into rat soleus, before BoNT/A administration, was insufficient to attenuate atrophy. It was also insufficient to decrease BoNT/A-induced Foxo activity at 3 days, although NF-κB activity was abolished. By 10 days both NF-κB and Foxo activation were abolished by Hsp70. Hsp70-overexpression was unable to alter the levels of BoNT/A-induced effects on MuRF1/atrogin-1, polyubiquitination, or proteasome activity. In conclusion, Hsp70 overexpression is insufficient to attenuate BoNT/A-induced atrophy. It remains unclear what proteolytic mechanism/s are contributing to BoNT/A-induced atrophy, although a Foxo-MuRF1-ubiquitin-proteasome contribution may exist, at least in early BoNT/A-induced atrophy. Further clarification of UPS involvement in BoNT/A-induced atrophy is warranted.
Collapse
Affiliation(s)
- Fraser E Houston
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida; and
| | - Brian A Hain
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida; and
| | - Thomas J Adams
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida; and
| | - Kati L Houston
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida; and
| | | | - Stephen L Dodd
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida; and
| |
Collapse
|
54
|
Abstract
Atrophy occurs in specific muscles with inactivity (for example, during plaster cast immobilization) or denervation (for example, in patients with spinal cord injuries). Muscle wasting occurs systemically in older people (a condition known as sarcopenia); as a physiological response to fasting or malnutrition; and in many diseases, including chronic obstructive pulmonary disorder, cancer-associated cachexia, diabetes, renal failure, cardiac failure, Cushing syndrome, sepsis, burns and trauma. The rapid loss of muscle mass and strength primarily results from excessive protein breakdown, which is often accompanied by reduced protein synthesis. This loss of muscle function can lead to reduced quality of life, increased morbidity and mortality. Exercise is the only accepted approach to prevent or slow atrophy. However, several promising therapeutic agents are in development, and major advances in our understanding of the cellular mechanisms that regulate the protein balance in muscle include the identification of several cytokines, particularly myostatin, and a common transcriptional programme that promotes muscle wasting. Here, we discuss these new insights and the rationally designed therapies that are emerging to combat muscle wasting.
Collapse
|
55
|
Zhao Q, Yang ST, Wang JJ, Zhou J, Xing SS, Shen CC, Wang XX, Yue YX, Song J, Chen M, Wei YY, Zhou QP, Dai T, Song YH. TNF alpha inhibits myogenic differentiation of C2C12 cells through NF-κB activation and impairment of IGF-1 signaling pathway. Biochem Biophys Res Commun 2015; 458:790-5. [PMID: 25686491 DOI: 10.1016/j.bbrc.2015.02.026] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 02/05/2015] [Indexed: 01/07/2023]
Abstract
Cachexia or muscle wasting is a common condition that occurs in many chronic diseases. The wasting conditions are characterized by increased levels of TNF-α which was also known as cachectin in the past. But how TNF-α exerts its cachetic effects remains controversial. To clarify this issue, we investigated the impact of TNF-α on C2C12 cell myogenic differentiation. Our results demonstrate that myotube formation was completely inhibited by TNF-α when added to differentiating C2C12 myoblasts. The inhibitory effect of TNF-α on differentiation was accompanied by activation of NF-κB and down regulation of myogenin and Akt. Importantly, TNF-α's effect on differentiation was abolished when IGF-1 was added to the culture. IGF-1 treatment also inhibited NF-κB reporter activity and restored Akt levels. Our data suggest that TNF-α inhibits myogenic differentiation through NF-κB activation and impairment of IGF-1 signaling pathway. The reversal of TNF-α induced inhibition of myogenesis by IGF-1 may have significant therapeutic potential.
Collapse
Affiliation(s)
- Q Zhao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - S T Yang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - J J Wang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China; The Affiliated Wuxi Hospital for Maternal and Child Health Care of Medical University of Nanjing, Wuxi, Jiangsu Province, China
| | - J Zhou
- Department of Surgery, First Affiliated Hospital, Soochow University, Suzhou, China
| | - S S Xing
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - C C Shen
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - X X Wang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Y X Yue
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - J Song
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - M Chen
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Y Y Wei
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Q P Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - T Dai
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Y H Song
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
56
|
Ulanova A, Gritsyna Y, Vikhlyantsev I, Salmov N, Bobylev A, Abdusalamova Z, Rogachevsky V, Shenkman B, Podlubnaya Z. Isoform composition and gene expression of thick and thin filament proteins in striated muscles of mice after 30-day space flight. BIOMED RESEARCH INTERNATIONAL 2015; 2015:104735. [PMID: 25664316 PMCID: PMC4312622 DOI: 10.1155/2015/104735] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 12/11/2014] [Accepted: 12/20/2014] [Indexed: 11/18/2022]
Abstract
Changes in isoform composition, gene expression of titin and nebulin, and isoform composition of myosin heavy chains as well as changes in titin phosphorylation level in skeletal (m. gastrocnemius, m. tibialis anterior, and m. psoas) and cardiac muscles of mice were studied after a 30-day-long space flight onboard the Russian spacecraft "BION-M" number 1. A muscle fibre-type shift from slow-to-fast and a decrease in the content of titin and nebulin in the skeletal muscles of animals from "Flight" group was found. Using Pro-Q Diamond staining, an ~3-fold increase in the phosphorylation level of titin in m. gastrocnemius of mice from the "Flight" group was detected. The content of titin and its phosphorylation level in the cardiac muscle of mice from "Flight" and "Control" groups did not differ; nevertheless an increase (2.2 times) in titin gene expression in the myocardium of flight animals was found. The observed changes are discussed in the context of their role in the contractile activity of striated muscles of mice under conditions of weightlessness.
Collapse
Affiliation(s)
- Anna Ulanova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino 142290, Russia
- Pushchino State Institute of Natural Science, Nauki Street 3, Pushchino 142290, Russia
| | - Yulia Gritsyna
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino 142290, Russia
| | - Ivan Vikhlyantsev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino 142290, Russia
| | - Nikolay Salmov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino 142290, Russia
| | - Alexander Bobylev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino 142290, Russia
| | - Zarema Abdusalamova
- Dagestan State University, Gadzhieva Street 43a, Makhachkala, Republic of Dagestan 367000, Russia
| | - Vadim Rogachevsky
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino 142290, Russia
| | - Boris Shenkman
- SRC, Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Street 76A, Moscow 123007, Russia
| | - Zoya Podlubnaya
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino 142290, Russia
- Pushchino State Institute of Natural Science, Nauki Street 3, Pushchino 142290, Russia
| |
Collapse
|
57
|
Maffei M, Longa E, Qaisar R, Agoni V, Desaphy JF, Camerino DC, Bottinelli R, Canepari M. Actin sliding velocity on pure myosin isoforms from hindlimb unloaded mice. Acta Physiol (Oxf) 2014; 212:316-29. [PMID: 24888432 DOI: 10.1111/apha.12320] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/07/2014] [Accepted: 05/25/2014] [Indexed: 11/29/2022]
Abstract
AIM Notwithstanding the widely accepted idea that following disuse skeletal muscles become faster, an increase in shortening velocity was previously observed mostly in fibres containing type 1 myosin, whereas a decrease was generally found in fibres containing type 2B myosin. In this study, unloaded shortening velocity of pure type 1 and 2B fibres from hindlimb unloaded mice was determined and a decrease in type 2B fibres was found. METHODS To clarify whether the decrease in shortening velocity could depend on alterations of myosin motor function, an in vitro motility assay approach was applied to study pure type 1 and pure type 2B myosin from hindlimb unloaded mice. The latter approach, assessing actin sliding velocity on isolated myosin in the absence of other myofibrillar proteins, enabled to directly investigate myosin motor function. RESULTS Actin sliding velocity was significantly lower on type 2B myosin following unloading (2.70 ± 0.32 μm s(-1)) than in control conditions (4.11 ± 0.35 μm s(-1)), whereas actin sliding velocity of type 1 myosin was not different following unloading (0.89 ± 0.04 μm s(-1)) compared with control conditions (0.84 ± 0.17 μm s(-1)). Myosin light chain (MLC) isoform composition of type 2B myosin from hindlimb unloaded and control mice was not different. No oxidation of either type 1 or 2B myosin was observed. Higher phosphorylation of regulatory MLC in type 2B myosin after unloading was found. CONCLUSION Results suggest that the observed lower shortening velocity of type 2B fibres following unloading could be related to slowing of acto-myosin kinetics in the presence of MLC phosphorylation.
Collapse
Affiliation(s)
- M. Maffei
- Department of Molecular Medicine and Interuniversity; Institute of Myology; University of Pavia; Pavia Italy
| | - E. Longa
- Department of Molecular Medicine and Interuniversity; Institute of Myology; University of Pavia; Pavia Italy
| | - R. Qaisar
- Department of Molecular Medicine and Interuniversity; Institute of Myology; University of Pavia; Pavia Italy
| | - V. Agoni
- Department of Molecular Medicine and Interuniversity; Institute of Myology; University of Pavia; Pavia Italy
| | - J.-F. Desaphy
- Section of Pharmacology; Department of Pharmacy and Drug Sciences and Interuniversity Institute of Myology; University of Bari - Aldo Moro; Bari Italy
| | - D. Conte Camerino
- Section of Pharmacology; Department of Pharmacy and Drug Sciences and Interuniversity Institute of Myology; University of Bari - Aldo Moro; Bari Italy
| | - R. Bottinelli
- Department of Molecular Medicine and Interuniversity; Institute of Myology; University of Pavia; Pavia Italy
- Fondazione Salvatore Maugeri (IRCCS); Scientific Institute of Pavia; Pavia Italy
- Interdipartimental Centre of Biology and Sport Medicine; University of Pavia; Pavia Italy
| | - M. Canepari
- Department of Molecular Medicine and Interuniversity; Institute of Myology; University of Pavia; Pavia Italy
| |
Collapse
|
58
|
Recent advances in mitochondrial turnover during chronic muscle disuse. Integr Med Res 2014; 3:161-171. [PMID: 28664093 PMCID: PMC5481769 DOI: 10.1016/j.imr.2014.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/06/2014] [Indexed: 12/21/2022] Open
Abstract
Chronic muscle disuse, such as that resulting from immobilization, denervation, or prolonged physical inactivity, produces atrophy and a loss of mitochondria, yet the molecular relationship between these events is not fully understood. In this review we attempt to identify the key regulatory steps mediating the loss of muscle mass and the decline in mitochondrial content and function. An understanding of common intracellular signaling pathways may provide much-needed insight into the possible therapeutic targets for treatments that will maintain aerobic energy metabolism and preserve muscle mass during disuse conditions.
Collapse
|
59
|
Blanqué R, Lepescheux L, Auberval M, Minet D, Merciris D, Cottereaux C, Clément-Lacroix P, Delerive P, Namour F. Characterization of GLPG0492, a selective androgen receptor modulator, in a mouse model of hindlimb immobilization. BMC Musculoskelet Disord 2014; 15:291. [PMID: 25185887 PMCID: PMC4167280 DOI: 10.1186/1471-2474-15-291] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 08/27/2014] [Indexed: 11/23/2022] Open
Abstract
Background Muscle wasting is a hallmark of many chronic conditions but also of aging and results in a progressive functional decline leading ultimately to disability. Androgens, such as testosterone were proposed as therapy to counteract muscle atrophy. However, this treatment is associated with potential cardiovascular and prostate cancer risks and therefore not acceptable for long-term treatment. Selective Androgen receptor modulators (SARM) are androgen receptor ligands that induce muscle anabolism while having reduced effects in reproductive tissues. Therefore, they represent an alternative to testosterone therapy. Our objective was to demonstrate the activity of SARM molecule (GLPG0492) on a immobilization muscle atrophy mouse model as compared to testosterone propionate (TP) and to identify putative biomarkers in the plasma compartment that might be related to muscle function and potentially translated into the clinical space. Methods GLPG0492, a non-steroidal SARM, was evaluated and compared to TP in a mouse model of hindlimb immobilization. Results GLPG0492 treatment partially prevents immobilization-induced muscle atrophy with a trend to promote muscle fiber hypertrophy in a dose-dependent manner. Interestingly, GLPG0492 was found as efficacious as TP at reducing muscle loss while sparing reproductive tissues. Furthermore, gene expression studies performed on tibialis samples revealed that both GLPG0492 and TP were slowing down muscle loss by negatively interfering with major signaling pathways controlling muscle mass homeostasis. Finally, metabolomic profiling experiments using 1H-NMR led to the identification of a plasma GLPG0492 signature linked to the modulation of cellular bioenergetic processes. Conclusions Taken together, these results unveil the potential of GLPG0492, a non-steroidal SARM, as treatment for, at least, musculo-skeletal atrophy consecutive to coma, paralysis, or limb immobilization. Electronic supplementary material The online version of this article (doi:10.1186/1471-2474-15-291) contains supplementary material, which is available to authorized users.
Collapse
|
60
|
Current understanding of sarcopenia: possible candidates modulating muscle mass. Pflugers Arch 2014; 467:213-29. [PMID: 24797147 DOI: 10.1007/s00424-014-1527-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 12/17/2022]
Abstract
The world's elderly population is expanding rapidly, and we are now faced with the significant challenge of maintaining or improving physical activity, independence, and quality of life in the elderly. Sarcopenia, the age-related loss of skeletal muscle mass, is characterized by a deterioration of muscle quantity and quality leading to a gradual slowing of movement, a decline in strength and power, increased risk of fall-related injury, and often, frailty. Since sarcopenia is largely attributed to various molecular mediators affecting fiber size, mitochondrial homeostasis, and apoptosis, the mechanisms responsible for these deleterious changes present numerous therapeutic targets for drug discovery. Muscle loss has been linked with several proteolytic systems, including the ubuiquitin-proteasome, lysosome-autophagy, and tumor necrosis factor (TNF)-α/nuclear factor-kappaB (NF-κB) systems. Although many factors are considered to regulate age-dependent muscle loss, this gentle atrophy is not affected by factors known to enhance rapid atrophy (denervation, hindlimb suspension, etc.). In addition, defects in Akt-mammalian target of rapamycin (mTOR) and serum response factor (SRF)-dependent signaling have been found in sarcopenic muscle. Intriguingly, more recent studies indicated an apparent functional defect in autophagy- and myostatin-dependent signaling in sarcopenic muscle. In this review, we summarize the current understanding of the adaptation of many regulators in sarcopenia.
Collapse
|
61
|
Montilla SIR, Johnson TP, Pearce SC, Gardan-Salmon D, Gabler NK, Ross JW, Rhoads RP, Baumgard LH, Lonergan SM, Selsby JT. Heat stress causes oxidative stress but not inflammatory signaling in porcine skeletal muscle. Temperature (Austin) 2014; 1:42-50. [PMID: 27583280 PMCID: PMC4972518 DOI: 10.4161/temp.28844] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 04/08/2014] [Accepted: 04/08/2014] [Indexed: 02/07/2023] Open
Abstract
Heat stress is associated with death and other maladaptions including muscle dysfunction and impaired growth across species. Despite this common observation, the molecular effects leading to these pathologic changes remain unclear. The purpose of this study was to determine the extent to which heat stress disrupted redox balance and initiated an inflammatory response in oxidative and glycolytic skeletal muscle. Female pigs (5-6/group) were subjected to thermoneutral (20 °C) or heat stress (35 °C) conditions for 1 or 3 days and the semitendinosus removed and dissected into red (STR) and white (STW) portions. After 1 day of heat stress, relative abundance of proteins modified by malondialdehyde, a measure of oxidative damage, was increased 2.5-fold (P < 0.05) compared with thermoneutral in the STR but not the STW, before returning to thermoneutral conditions following 3 days of heat stress. This corresponded with increased catalase and superoxide dismutase-1 gene expression (P < 0.05) and superoxide dismutase-1 protein abundance (P < 0.05) in the STR but not the STW. In the STR catalase and total superoxide dismutase activity were increased by ~30% and ~130%, respectively (P < 0.05), after 1 day of heat stress and returned to thermoneutral levels by day 3. One or 3 days of heat stress did not increase inflammatory signaling through the NF-κB pathway in the STR or STW. These data suggest that oxidative muscle is more susceptible to heat stress-mediated changes in redox balance than glycolytic muscle during chronic heat stress.
Collapse
Affiliation(s)
| | | | - Sarah C Pearce
- Department of Animal Science; Iowa State University; Ames, IA USA
| | | | | | - Jason W Ross
- Department of Animal Science; Iowa State University; Ames, IA USA
| | - Robert P Rhoads
- Department of Animal and Poultry Sciences; Virginia Tech; Blacksburg, VA USA
| | - Lance H Baumgard
- Department of Animal Science; Iowa State University; Ames, IA USA
| | | | - Joshua T Selsby
- Department of Animal Science; Iowa State University; Ames, IA USA
| |
Collapse
|
62
|
Wu CL, Cornwell EW, Jackman RW, Kandarian SC. NF-κB but not FoxO sites in the MuRF1 promoter are required for transcriptional activation in disuse muscle atrophy. Am J Physiol Cell Physiol 2014; 306:C762-7. [PMID: 24553183 DOI: 10.1152/ajpcell.00361.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The muscle-specific ring finger protein 1 (MuRF1) gene is required for most types of skeletal muscle atrophy yet we have little understanding of its transcriptional regulation. The purpose of this study is to identify whether NF-κB and/or FoxO response elements in the MuRF1 promoter are required for MuRF1 gene activation during skeletal muscle atrophy due to the removal of hindlimb weight bearing ("unloading"). Both NF-κB -dependent and FoxO-dependent luciferase reporter activities were significantly increased at 5 days of unloading. Using a 4.4-kb MuRF1 promoter reporter construct, a fourfold increase in reporter (i.e., luciferase) activity was found in rat soleus muscles after 5 days of hindlimb unloading. This activation was abolished by mutagenesis of either of the two distal putative NF-κB sites or all three putative NF-κB sites but not by mutagenesis of all four putative FoxO sites. This work provides the first direct evidence that NF-κB sites, but not FoxO sites, are required for MuRF1 promoter activation in muscle disuse atrophy in vivo.
Collapse
Affiliation(s)
- Chia-Ling Wu
- Department of Health Sciences, Boston University, Boston, Massachusetts
| | | | | | | |
Collapse
|
63
|
Cornwell EW, Mirbod A, Wu CL, Kandarian SC, Jackman RW. C26 cancer-induced muscle wasting is IKKβ-dependent and NF-kappaB-independent. PLoS One 2014; 9:e87776. [PMID: 24489962 PMCID: PMC3906224 DOI: 10.1371/journal.pone.0087776] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/30/2013] [Indexed: 12/24/2022] Open
Abstract
Existing data suggest that NF-kappaB signaling is a key regulator of cancer-induced skeletal muscle wasting. However, identification of the components of this signaling pathway and of the NF-κB transcription factors that regulate wasting is far from complete. In muscles of C26 tumor bearing mice, overexpression of dominant negative (d.n.) IKKβ blocked muscle wasting by 69% and the IκBα-super repressor blocked wasting by 41%. In contrast, overexpression of d.n. IKKα or d.n. NIK did not block C26-induced wasting. Surprisingly, overexpression of d.n. p65 or d.n. c-Rel did not significantly affect muscle wasting. Genome-wide mRNA expression arrays showed upregulation of many genes previously implicated in muscle atrophy. To test if these upregulated genes were direct targets of NF-κB transcription factors, we compared genome-wide p65 binding to DNA in control and cachectic muscle using ChIP-sequencing. Bioinformatic analysis of ChIP-sequencing data from control and C26 muscles showed very little p65 binding to genes in cachexia and little to suggest that upregulated p65 binding influences the gene expression associated with muscle based cachexia. The p65 ChIP-seq data are consistent with our finding of no significant change in protein binding to an NF-κB oligonucleotide in a gel shift assay, no activation of a NF-κB-dependent reporter, and no effect of d.n.p65 overexpression in muscles of tumor bearing mice. Taken together, these data support the idea that although inhibition of IκBα, and particularly IKKβ, blocks cancer-induced wasting, the alternative NF-κB signaling pathway is not required. In addition, the downstream NF-κB transcription factors, p65 and c-Rel do not appear to regulate the transcriptional changes induced by the C26 tumor. These data are consistent with the growing body of literature showing that there are NF-κB-independent substrates of IKKβ and IκBα that regulate physiological processes.
Collapse
Affiliation(s)
- Evangeline W. Cornwell
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Azadeh Mirbod
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Chia-Ling Wu
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Susan C. Kandarian
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Robert W. Jackman
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
64
|
Donaldson A, Natanek SA, Lewis A, Man WDC, Hopkinson NS, Polkey MI, Kemp PR. Increased skeletal muscle-specific microRNA in the blood of patients with COPD. Thorax 2013; 68:1140-9. [PMID: 23814167 PMCID: PMC3841809 DOI: 10.1136/thoraxjnl-2012-203129] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 06/03/2013] [Accepted: 06/06/2013] [Indexed: 01/23/2023]
Abstract
BACKGROUND Skeletal muscle weakness in chronic obstructive pulmonary disease (COPD) carries a poor prognosis, therefore a non-invasive marker of this process could be useful. Reduced expression of muscle-specific microRNA (myomiRs) in quadriceps muscle in patients with COPD is associated with skeletal muscle weakness and changes in muscle fibre composition. Circulating exosomal miRNAs can be measured in blood, making them candidate biomarkers of biopsy phenotype. To determine whether plasma myomiR levels were associated with fibre size or fibre proportion, we measured myomiRs in plasma from patients with COPD and healthy controls. METHODS AND RESULTS 103 patients with COPD and 25 age-matched controls were studied. Muscle-specific miRNA was elevated in the plasma of patients with COPD and showed distinct patterns. Specifically, miR-1 was inversely associated with fat-free mass in the cohort, whereas levels of miR-499 were more directly associated with strength and quadriceps type I fibre proportion. Two miRs not restricted to muscle in origin (miR-16 and miR-122) did not differ between patients and controls. Plasma miR-499 was also associated with muscle nuclear factor κB p50 but not p65 in patients with early COPD whereas plasma inflammatory cytokines were associated with miR-206 in patients with more advanced disease. CONCLUSIONS Plasma levels of individual myomiRs are altered in patients with COPD but alone do not predict muscle fibre size or proportion. Our findings are consistent with an increase in muscle wasting and turnover associated with the development of skeletal muscle dysfunction and fibre-type shift in patients with stable COPD.
Collapse
Affiliation(s)
- Anna Donaldson
- Section of Molecular Medicine, National Heart and Lung Institute, Imperial College London, , London, UK
| | | | | | | | | | | | | |
Collapse
|
65
|
Abstract
Striated respiratory muscles are necessary for lung ventilation and to maintain the patency of the upper airway. The basic structural and functional properties of respiratory muscles are similar to those of other striated muscles (both skeletal and cardiac). The sarcomere is the fundamental organizational unit of striated muscles and sarcomeric proteins underlie the passive and active mechanical properties of muscle fibers. In this respect, the functional categorization of different fiber types provides a conceptual framework to understand the physiological properties of respiratory muscles. Within the sarcomere, the interaction between the thick and thin filaments at the level of cross-bridges provides the elementary unit of force generation and contraction. Key to an understanding of the unique functional differences across muscle fiber types are differences in cross-bridge recruitment and cycling that relate to the expression of different myosin heavy chain isoforms in the thick filament. The active mechanical properties of muscle fibers are characterized by the relationship between myoplasmic Ca2+ and cross-bridge recruitment, force generation and sarcomere length (also cross-bridge recruitment), external load and shortening velocity (cross-bridge cycling rate), and cross-bridge cycling rate and ATP consumption. Passive mechanical properties are also important reflecting viscoelastic elements within sarcomeres as well as the extracellular matrix. Conditions that affect respiratory muscle performance may have a range of underlying pathophysiological causes, but their manifestations will depend on their impact on these basic elemental structures.
Collapse
Affiliation(s)
- Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | | | | | | |
Collapse
|
66
|
Xiang J, Zhao Y, Chen J, Zhou J. Expression of basic fibroblast growth factor, protein kinase C and members of the apoptotic pathway in skeletal muscle of streptozotocin-induced diabetic rats. Tissue Cell 2013; 46:1-8. [PMID: 24008114 DOI: 10.1016/j.tice.2013.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/22/2013] [Accepted: 07/22/2013] [Indexed: 01/11/2023]
Abstract
This study investigated the potential mechanisms that may underlie diabetes induced amyoatrophy. Sprague-Dawley rats were either injected intraperiotneally with STZ (test group; N=8) to induce diabetic-like symptoms (blood glucose level ≥16.65mmol/L) or with buffer (control group; N=8). Differences in muscle structure between the STZ-induced diabetic and control groups were evaluated by histochemistry. Protein and mRNA levels of basic FGF (bFGF), bax, bcl-2, and caspase 3 in skeletal muscle were compared between the 2 groups using immunohistochemistry and quantitative PCR, respectively. Serum level of insulin and protein kinase C (PKC) were measured by competitive RIA and ELISA, respectively. Unlike control animals, the skeletal muscle fibers from STZ-induced diabetic animals were broken and pyknotic, the sarcomeric structure disrupted, and mild hyperplasia of interstitial adipose tissues was detected. The serum level of PKC was higher (P=0.003) and the protein and mRNA levels of bFGF in skeletal muscle were lower (P=0.001) in STZ-induced diabetic versus control animals. Protein and mRNA levels of the apoptosis promoting genes caspase-3 and bax were higher in skeletal muscle from STZ-induced diabetic rats as compared to control animals (P<0.001 and P=0.037, respectively), while mRNA and protein levels of bcl-2, an inhibitor of apoptosis, was lower in STZ-induced diabetic rats versus control animals (P=0.026). Increasing apoptosis in skeletal muscle from STZ-induced diabetic rats was further demonstrated by TNNEL assay. Our findings suggest that enhanced PKC levels, reduction of bFGF expression, and increased in apoptosis might be associated with the development of diabetes-induced myoatrophy.
Collapse
Affiliation(s)
- Jingyan Xiang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Jingjiong Chen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jian Zhou
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
67
|
Pond AL, Nedele C, Wang WH, Wang X, Walther C, Jaeger C, Bradley KS, Du H, Fujita N, Hockerman GH, Hannon KM. The mERG1a channel modulates skeletal muscle MuRF1, but not MAFbx, expression. Muscle Nerve 2013; 49:378-88. [PMID: 23761265 DOI: 10.1002/mus.23924] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2013] [Indexed: 12/31/2022]
Abstract
INTRODUCTION We investigated the mechanism by which the MERG1a K+ channel increases ubiquitin proteasome proteolysis (UPP). METHODS Hindlimb suspension and electro-transfer of Merg1a cDNA into mouse gastrocnemius muscles induced atrophy. RESULTS Atrophic gastrocnemius muscles of hindlimb-suspended mice express Merg1a, Murf1, and Mafbx genes. Electrotransfer of Merg1a significantly decreases muscle fiber size (12.6%) and increases UPP E3 ligase Murf1 mRNA (2.1-fold) and protein (23.7%), but does not affect Mafbx E3 ligase expression. Neither Merg1a-induced decreased fiber size nor Merg1a-induced increased Murf1 expression is curtailed significantly by coexpression of inactive HR-Foxo3a, a gene encoding a transcription factor known to induce Mafbx expression. CONCLUSIONS The MERG1a K+ channel significantly increases expression of Murf1, but not Mafbx. We explored this expression pattern by expressing inactive Foxo3a and showing that it is not involved in MERG1a-mediated expression of Murf1. These findings suggest that MERG1a may not modulate Murf1 expression through the AKT/FOXO pathway.
Collapse
Affiliation(s)
- Amber L Pond
- Anatomy Department, Southern Illinois University School of Medicine, 2080 Life Sciences III, 1135 Lincoln Drive, Carbondale, Illinois, 62901, USA; Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy, Purdue University, West Lafayette, Indiana, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Bodine SC. Disuse-induced muscle wasting. Int J Biochem Cell Biol 2013; 45:2200-8. [PMID: 23800384 DOI: 10.1016/j.biocel.2013.06.011] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 06/11/2013] [Accepted: 06/12/2013] [Indexed: 12/17/2022]
Abstract
Loss of skeletal muscle mass occurs frequently in clinical settings in response to joint immobilization and bed rest, and is induced by a combination of unloading and inactivity. Disuse-induced atrophy will likely affect every person in his or her lifetime, and can be debilitating especially in the elderly. Currently there are no good therapies to treat disuse-induced muscle atrophy, in part, due to a lack of understanding of the cellular and molecular mechanisms responsible for the induction and maintenance of muscle atrophy. Our current understanding of disuse atrophy comes from the investigation of a variety of models (joint immobilization, hindlimb unloading, bed rest, spinal cord injury) in both animals and humans. Under conditions of unloading, it is widely accepted that there is a decrease in protein synthesis, however, the role of protein degradation, especially in humans, is debated. This review will examine the current understanding of the molecular and cellular mechanisms regulating muscle loss under disuse conditions, discussing the similarities and areas of dispute between the animal and human literature. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- Sue C Bodine
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, One Shields Avenue, Davis, CA 95616, United States.
| |
Collapse
|
69
|
Bonaldo P, Sandri M. Cellular and molecular mechanisms of muscle atrophy. Dis Model Mech 2013; 6:25-39. [PMID: 23268536 PMCID: PMC3529336 DOI: 10.1242/dmm.010389] [Citation(s) in RCA: 862] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle is a plastic organ that is maintained by multiple pathways regulating cell and protein turnover. During muscle atrophy, proteolytic systems are activated, and contractile proteins and organelles are removed, resulting in the shrinkage of muscle fibers. Excessive loss of muscle mass is associated with poor prognosis in several diseases, including myopathies and muscular dystrophies, as well as in systemic disorders such as cancer, diabetes, sepsis and heart failure. Muscle loss also occurs during aging. In this paper, we review the key mechanisms that regulate the turnover of contractile proteins and organelles in muscle tissue, and discuss how impairments in these mechanisms can contribute to muscle atrophy. We also discuss how protein synthesis and degradation are coordinately regulated by signaling pathways that are influenced by mechanical stress, physical activity, and the availability of nutrients and growth factors. Understanding how these pathways regulate muscle mass will provide new therapeutic targets for the prevention and treatment of muscle atrophy in metabolic and neuromuscular diseases.
Collapse
Affiliation(s)
- Paolo Bonaldo
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy.
| | | |
Collapse
|
70
|
Nakamura H, Aoki K, Masuda W, Alles N, Nagano K, Fukushima H, Osawa K, Yasuda H, Nakamura I, Mikuni-Takagaki Y, Ohya K, Maki K, Jimi E. Disruption of NF-κB1 prevents bone loss caused by mechanical unloading. J Bone Miner Res 2013; 28:1457-67. [PMID: 23322687 DOI: 10.1002/jbmr.1866] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 11/28/2012] [Accepted: 12/18/2012] [Indexed: 02/03/2023]
Abstract
Mechanical unloading, such as in a microgravity environment in space or during bed rest (for patients who require prolonged bed rest), leads to a decrease in bone mass because of the suppression of bone formation and the stimulation of bone resorption. To address the challenges presented by a prolonged stay in space and the forthcoming era of a super-aged society, it will be important to prevent the bone loss caused by prolonged mechanical unloading. Nuclear factor κB (NF-κB) transcription factors are activated by mechanical loading and inflammatory cytokines. Our objective was to elucidate the role of NF-κB pathways in bone loss that are caused by mechanical unloading. Eight-week-old wild-type (WT) and NF-κB1-deficient mice were randomly assigned to a control or mechanically unloaded with tail suspension group. After 2 weeks, a radiographic analysis indicated a decrease in bone mass in the tibias and femurs of the unloaded WT mice but not in the NF-κB1-deficient mice. An NF-κB1 deficiency suppressed the unloading-induced reduction in bone formation by maintaining the proportion and/or potential of osteoprogenitors or immature osteoblasts, and by suppression of bone resorption through the inhibition of intracellular signaling through the receptor activator of NF-κB ligand (RANKL) in osteoclast precursors. Thus, NF-κB1 is involved in two aspects of rapid reduction in bone mass that are induced by disuse osteoporosis in space or bed rest.
Collapse
Affiliation(s)
- Hitomi Nakamura
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Song YH, Song JL, Delafontaine P, Godard MP. The therapeutic potential of IGF-I in skeletal muscle repair. Trends Endocrinol Metab 2013; 24:310-9. [PMID: 23628587 PMCID: PMC3732824 DOI: 10.1016/j.tem.2013.03.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/22/2013] [Accepted: 03/22/2013] [Indexed: 12/30/2022]
Abstract
Skeletal muscle loss due to aging, motor-neuron degeneration, cancer, heart failure, and ischemia is a serious condition for which currently there is no effective treatment. Insulin-like growth factor 1 (IGF-I) plays an important role in muscle maintenance and repair. Preclinical studies have shown that IGF-I is involved in increasing muscle mass and strength, reducing degeneration, inhibiting the prolonged and excessive inflammatory process due to toxin injury, and increasing the proliferation potential of satellite cells. However, clinical trials have not been successful due to ineffective delivery methods. Choosing the appropriate isoforms or peptides and developing targeted delivery techniques can resolve this issue. Here we discuss the latest development in the field with special emphasis on novel therapeutic approaches.
Collapse
Affiliation(s)
- Yao-Hua Song
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China
- Corresponding authors: Yao-Hua Song, M.D. Ph.D., Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China, Phone: 86-512-65880899/626, Fax: 86-512-65880929,
| | - Jenny L. Song
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China
| | - Patrice Delafontaine
- Tulane University Heart and Vascular Institute, Tulane University School of Medicine
- Corresponding authors: Yao-Hua Song, M.D. Ph.D., Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China, Phone: 86-512-65880899/626, Fax: 86-512-65880929,
| | - Michael P. Godard
- Department of Nutrition and Kinesiology, University of Central Missouri, Warrensburg, MO
- Corresponding authors: Yao-Hua Song, M.D. Ph.D., Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, 199 Ren Ai Road, Suzhou 215123, China, Phone: 86-512-65880899/626, Fax: 86-512-65880929,
| |
Collapse
|
72
|
Abstract
PURPOSE OF REVIEW There are a variety of pathophysiologic conditions that are known to induce skeletal muscle atrophy. However, muscle wasting can occur through multiple distinct signaling pathways with differential sensitivity between selective skeletal muscle fiber subtypes. This review summarizes some of the underlying molecular mechanisms responsible for fiber-specific muscle mass regulation. RECENT FINDINGS Peroxisome proliferator-activated receptor gamma coactivator 1-alpha protects slow-twitch oxidative fibers from denervation/immobilization (disuse)-induced muscle atrophies. Nutrient-related muscle atrophies, such as those induced by cancer cachexia, sepsis, chronic heart failure, or diabetes, are largely restricted to fast-twitch glycolytic fibers, of which the underlying mechanism is usually related to abnormality of protein degradation, including proteasomal and lysosomal pathways. In contrast, nuclear factor kappaB activation apparently serves a dual function by inducing both fast-twitch fiber atrophy and slow-twitch fiber degeneration. SUMMARY Fast-twitch glycolytic fibers are more vulnerable than slow-twitch oxidative fibers under a variety of atrophic conditions related to signaling transduction of Forkhead box O family, autophagy inhibition, transforming growth factor beta family, and nuclear factor-kappaB. The resistance of oxidative fibers may result from the protection of peroxisome proliferator-activated receptor gamma coactivator 1-alpha.
Collapse
Affiliation(s)
- Yichen Wang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Diabetes Research and Training Center, Bronx, New York, USA
| | - Jeffrey E. Pessin
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Diabetes Research and Training Center, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Diabetes Research and Training Center, Bronx, New York, USA
| |
Collapse
|
73
|
Schiaffino S, Dyar KA, Ciciliot S, Blaauw B, Sandri M. Mechanisms regulating skeletal muscle growth and atrophy. FEBS J 2013; 280:4294-314. [PMID: 23517348 DOI: 10.1111/febs.12253] [Citation(s) in RCA: 978] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/13/2013] [Accepted: 03/14/2013] [Indexed: 12/11/2022]
Abstract
Skeletal muscle mass increases during postnatal development through a process of hypertrophy, i.e. enlargement of individual muscle fibers, and a similar process may be induced in adult skeletal muscle in response to contractile activity, such as strength exercise, and specific hormones, such as androgens and β-adrenergic agonists. Muscle hypertrophy occurs when the overall rates of protein synthesis exceed the rates of protein degradation. Two major signaling pathways control protein synthesis, the IGF1-Akt-mTOR pathway, acting as a positive regulator, and the myostatin-Smad2/3 pathway, acting as a negative regulator, and additional pathways have recently been identified. Proliferation and fusion of satellite cells, leading to an increase in the number of myonuclei, may also contribute to muscle growth during early but not late stages of postnatal development and in some forms of muscle hypertrophy in the adult. Muscle atrophy occurs when protein degradation rates exceed protein synthesis, and may be induced in adult skeletal muscle in a variety of conditions, including starvation, denervation, cancer cachexia, heart failure and aging. Two major protein degradation pathways, the proteasomal and the autophagic-lysosomal pathways, are activated during muscle atrophy and variably contribute to the loss of muscle mass. These pathways involve a variety of atrophy-related genes or atrogenes, which are controlled by specific transcription factors, such as FoxO3, which is negatively regulated by Akt, and NF-κB, which is activated by inflammatory cytokines.
Collapse
|
74
|
Radak Z, Zhao Z, Koltai E, Ohno H, Atalay M. Oxygen consumption and usage during physical exercise: the balance between oxidative stress and ROS-dependent adaptive signaling. Antioxid Redox Signal 2013; 18:1208-46. [PMID: 22978553 PMCID: PMC3579386 DOI: 10.1089/ars.2011.4498] [Citation(s) in RCA: 401] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The complexity of human DNA has been affected by aerobic metabolism, including endurance exercise and oxygen toxicity. Aerobic endurance exercise could play an important role in the evolution of Homo sapiens, and oxygen was not important just for survival, but it was crucial to redox-mediated adaptation. The metabolic challenge during physical exercise results in an elevated generation of reactive oxygen species (ROS) that are important modulators of muscle contraction, antioxidant protection, and oxidative damage repair, which at moderate levels generate physiological responses. Several factors of mitochondrial biogenesis, such as peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α), mitogen-activated protein kinase, and SIRT1, are modulated by exercise-associated changes in the redox milieu. PGC-1α activation could result in decreased oxidative challenge, either by upregulation of antioxidant enzymes and/or by an increased number of mitochondria that allows lower levels of respiratory activity for the same degree of ATP generation. Endogenous thiol antioxidants glutathione and thioredoxin are modulated with high oxygen consumption and ROS generation during physical exercise, controlling cellular function through redox-sensitive signaling and protein-protein interactions. Endurance exercise-related angiogenesis, up to a significant degree, is regulated by ROS-mediated activation of hypoxia-inducible factor 1α. Moreover, the exercise-associated ROS production could be important to DNA methylation and post-translation modifications of histone residues, which create heritable adaptive conditions based on epigenetic features of chromosomes. Accumulating data indicate that exercise with moderate intensity has systemic and complex health-promoting effects, which undoubtedly involve regulation of redox homeostasis and signaling.
Collapse
Affiliation(s)
- Zsolt Radak
- Faculty of Physical Education and Sport Science, Institute of Sport Science, Semmelweis University, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
75
|
Romanick M, Thompson LV, Brown-Borg HM. Murine models of atrophy, cachexia, and sarcopenia in skeletal muscle. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1410-20. [PMID: 23523469 DOI: 10.1016/j.bbadis.2013.03.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/10/2013] [Accepted: 03/12/2013] [Indexed: 12/11/2022]
Abstract
With the extension of life span over the past several decades, the age-related loss of muscle mass and strength that characterizes sarcopenia is becoming more evident and thus, has a more significant impact on society. To determine ways to intervene and delay, or even arrest the physical frailty and dependence that accompany sarcopenia, it is necessary to identify those biochemical pathways that define this process. Animal models that mimic one or more of the physiological pathways involved with this phenomenon are very beneficial in providing an understanding of the cellular processes at work in sarcopenia. The ability to influence pathways through genetic manipulation gives insight into cellular responses and their impact on the physical expression of sarcopenia. This review evaluates several murine models that have the potential to elucidate biochemical processes integral to sarcopenia. Identifying animal models that reflect sarcopenia or its component pathways will enable researchers to better understand those pathways that contribute to age-related skeletal muscle mass loss, and in turn, develop interventions that will prevent, retard, arrest, or reverse this phenomenon. This article is part of a Special Issue entitled: Animal Models of Disease.
Collapse
Affiliation(s)
- Mark Romanick
- Department of Physical Therapy, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | | | | |
Collapse
|
76
|
Abstract
PURPOSE OF REVIEW ICU-acquired weakness (ICUAW) is now recognized as a major complication of critical illness. There is no doubt that ICUAW is prevalent - some might argue ubiquitous - after critical illness, but its true role, the interaction with preexisting nerve and muscle lesions as well as its contribution to long-term functional disability, remains to be elucidated. RECENT FINDINGS In this article, we review the current state-of-the-art of the basic pathophysiology of nerve and muscle weakness after critical illness and explore the current literature on ICUAW with a special emphasis on the most important mechanisms of weakness. SUMMARY Variable contributions of structural and functional changes likely contribute to both early and late myopathy and neuropathy, although the specifics of the temporality of both processes, and the influence patient comorbidities, age, and nature of the ICU insult have on them, remain to be determined.
Collapse
|
77
|
Kostrominova TY, Reiner DS, Haas RH, Ingermanson R, McDonough PM. Automated methods for the analysis of skeletal muscle fiber size and metabolic type. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 306:275-332. [PMID: 24016528 DOI: 10.1016/b978-0-12-407694-5.00007-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
It is of interest to quantify the size, shape, and metabolic subtype of skeletal muscle fibers in many areas of biomedical research. To do so, skeletal muscle samples are sectioned transversely to the length of the muscle and labeled for extracellular or membrane proteins to delineate the fiber boundaries and additionally for biomarkers related to function or metabolism. The samples are digitally photographed and the fibers "outlined" for quantification of fiber cross-sectional area (CSA) using pointing devices interfaced to a computer, which is tedious, prone to error, and can be nonobjective. Here, we review methods for characterizing skeletal muscle fibers and describe new automated techniques, which rapidly quantify CSA and biomarkers. We discuss the applications of these methods to the characterization of mitochondrial dysfunctions, which underlie a variety of human afflictions, and we present a novel approach, utilizing images from the online Human Protein Atlas to predict relationships between fiber-specific protein expression, function, and metabolism.
Collapse
|
78
|
Okuneva AD, Vikhlyantsev IM, Shpagina MD, Rogachevskii VV, Khutzyan SS, Podlubnaya ZA, Grigoriev AI. Changes in titin and myosin heavy chain isoform composition in skeletal muscles of Mongolian Gerbil (Meriones unguiculatus) after 12-day spaceflight. Biophysics (Nagoya-shi) 2012. [DOI: 10.1134/s0006350912050144] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
79
|
Jackman RW, Wu CL, Kandarian SC. The ChIP-seq-defined networks of Bcl-3 gene binding support its required role in skeletal muscle atrophy. PLoS One 2012; 7:e51478. [PMID: 23251550 PMCID: PMC3519692 DOI: 10.1371/journal.pone.0051478] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 11/07/2012] [Indexed: 12/11/2022] Open
Abstract
NF-kappaB transcriptional activation is required for skeletal muscle disuse atrophy. We are continuing to study how the activation of NF-kB regulates the genes that encode the protein products that cause atrophy. Using ChIP-sequencing we found that Bcl-3, an NF-kB transcriptional activator required for atrophy, binds to the promoters of a number of genes whose collective function describes two major aspects of muscle wasting. By means of bioinformatics analysis of ChIP-sequencing data we found Bcl-3 to be directing transcription networks of proteolysis and energy metabolism. The proteolytic arm of the Bcl-3 networks includes many E3 ligases associated with proteasomal protein degradation, including that of the N-end rule pathway. The metabolic arm appears to be involved in organizing the change from oxidative phosphorylation to glycolysis in atrophying muscle. For one gene, MuRF1, ChIP-sequencing data identified the location of Bcl-3 and p50 binding in the promoter region which directed the creation of deletant and base-substitution mutations of MuRF1 promoter constructs to determine the effect on gene transcription. The results provide the first direct confirmation that the NF-kB binding site is involved in the muscle unloading regulation of MuRF1. Finally, we have combined the ChIP-sequencing results with gene expression microarray data from unloaded muscle to map several direct targets of Bcl-3 that are transcription factors whose own targets describe a set of indirect targets for NF-kB in atrophy. ChIP-sequencing provides the first molecular explanation for the finding that Bcl3 knockout mice are resistant to disuse muscle atrophy. Mapping the transcriptional regulation of muscle atrophy requires an unbiased analysis of the whole genome, which we show is now possible with ChIP-sequencing.
Collapse
Affiliation(s)
- Robert W Jackman
- Department of Health Sciences, Boston University, Boston, Massachusetts, USA.
| | | | | |
Collapse
|
80
|
Jackman RW, Cornwell EW, Wu CL, Kandarian SC. Nuclear factor-κB signalling and transcriptional regulation in skeletal muscle atrophy. Exp Physiol 2012; 98:19-24. [PMID: 22848079 DOI: 10.1113/expphysiol.2011.063321] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The nuclear factor-κB (NF-κB) signalling pathway is a necessary component of adult skeletal muscle atrophy resulting from systemic illnesses or disuse. Studies showing a role for the NF-κB pathway in muscle disuse include unloading, denervation and immobilization, and studies showing a role for NF-κB in systemic illnesses include cancer, chronic heart failure and acute septic lung injury. Muscle atrophy due to most of these triggers is associated with activation of NF-κB transcriptional activity. With the exception of muscle unloading, however, there is a paucity of data on the NF-κB transcription factors that regulate muscle atrophy, and little is known about which genes are targeted by NF-κB transcription factors during atrophy. Interestingly, in some cases it appears that the amelioration of muscle atrophy by genetic inhibition of NF-κB signalling proteins is due to effects that are independent of the downstream NF-κB transcription factors. These questions are prime areas for investigation if we are to understand a key component of muscle wasting in adult skeletal muscle.
Collapse
Affiliation(s)
- Robert W Jackman
- Boston University, Department of Health Sciences, 635 Commonwealth Avenue, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
81
|
Tonkin J, Villarroya F, Puri PL, Vinciguerra M. SIRT1 signaling as potential modulator of skeletal muscle diseases. Curr Opin Pharmacol 2012; 12:372-6. [DOI: 10.1016/j.coph.2012.02.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 02/03/2012] [Accepted: 02/15/2012] [Indexed: 12/20/2022]
|
82
|
Bhatnagar S, Mittal A, Gupta SK, Kumar A. TWEAK causes myotube atrophy through coordinated activation of ubiquitin-proteasome system, autophagy, and caspases. J Cell Physiol 2012; 227:1042-51. [PMID: 21567392 DOI: 10.1002/jcp.22821] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Proinflammatory cytokine TWEAK has now emerged as a key mediator of skeletal muscle-wasting in many catabolic conditions. However, the mechanisms by which TWEAK induces muscle proteolysis remain poorly understood. Here, we have investigated the role of ubiquitin-proteasome system, autophagy, and caspases in TWEAK-induced muscle wasting. Addition of TWEAK to C2C12 myotubes stimulated the ubiquitination of myosin heavy chain (MyHC) and augmented the expression of E3 ubiquitin ligase MuRF1. Pretreatment of myotubes with proteasome inhibitors MG132 or lactacystin or knockdown of MuRF1 by RNAi blocked the TWEAK-induced degradation of MyHC and myotube atrophy. TWEAK increased the expression of several autophagy-related molecules. Moreover, the inhibitors of autophagy improved the levels of MyHC in TWEAK-treated myotubes. TWEAK also increased activity of caspases in C2C12 myotubes. Pan-caspase or caspase 3 inhibitory peptide inhibited the TWEAK-induced loss of MyHC and myotube diameter. Our study demonstrates that nuclear factor-kappa B (NF-κB) transcription factor is essential for TWEAK-induced expression of MuRF1 and Beclin1. Furthermore, our results suggest that caspases contribute, at least in part, to the activation of NF-κB in response to TWEAK treatment. Collectively, the present study provides novel insight into the mechanisms of action of TWEAK in skeletal muscle.
Collapse
Affiliation(s)
- Shephali Bhatnagar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | | | | | | |
Collapse
|
83
|
Nuclear factor-κB signaling contributes to mechanical ventilation-induced diaphragm weakness*. Crit Care Med 2012; 40:927-34. [PMID: 22080641 DOI: 10.1097/ccm.0b013e3182374a84] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Although mechanical ventilation is a life-saving measure for patients in respiratory failure, prolonged mechanical ventilation results in diaphragmatic weakness attributable to fiber atrophy and contractile dysfunction. Therefore, identifying the signaling pathways responsible for mechanical ventilation-induced diaphragmatic weakness is important. In this context, it is established that oxidative stress is required for mechanical ventilation-induced diaphragmatic weakness to occur. Numerous redox-sensitive signaling pathways exist in muscle including the transcription factor nuclear factor-κB. Although it has been suggested that nuclear factor-κB contributes to proteolytic signaling in inactivity-induced atrophy in locomotor muscles, the role that nuclear factor-κB plays in mechanical ventilation-induced diaphragmatic weakness is unknown. We tested the hypothesis that nuclear factor-κB activation plays a key signaling role in mechanical ventilation-induced diaphragmatic weakness and that oxidative stress is required for nuclear factor-κB activation. DESIGN Cause and effect was determined by independently treating mechanically ventilated animals with either a specific nuclear factor-κB inhibitor (SN50) or a clinically relevant antioxidant (curcumin). MEASUREMENTS AND MAIN RESULTS Inhibition of nuclear factor-κB activity partially attenuated both mechanical ventilation-induced diaphragmatic atrophy and contractile dysfunction. Further, treatment with the antioxidant curcumin prevented mechanical ventilation-induced activation of nuclear factor-κB in the diaphragm and rescued the diaphragm from both mechanical ventilation-induced atrophy and contractile dysfunction. CONCLUSIONS Collectively, these findings support the hypothesis that nuclear factor-κB activation plays a significant signaling role in mechanical ventilation-induced diaphragmatic weakness and that oxidative stress is an upstream activator of nuclear factor-κB. Finally, our results suggest that prevention of mechanical ventilation-induced oxidative stress in the diaphragm could be a useful clinical strategy to prevent or delay mechanical ventilation-induced diaphragmatic weakness.
Collapse
|
84
|
Abstract
Muscle plasticity is defined as the ability of a given muscle to alter its structural and functional properties in accordance with the environmental conditions imposed on it. As such, respiratory muscle is in a constant state of remodeling, and the basis of muscle's plasticity is its ability to change protein expression and resultant protein balance in response to varying environmental conditions. Here, we will describe the changes of respiratory muscle imposed by extrinsic changes in mechanical load, activity, and innervation. Although there is a large body of literature on the structural and functional plasticity of respiratory muscles, we are only beginning to understand the molecular-scale protein changes that contribute to protein balance. We will give an overview of key mechanisms regulating protein synthesis and protein degradation, as well as the complex interactions between them. We suggest future application of a systems biology approach that would develop a mathematical model of protein balance and greatly improve treatments in a variety of clinical settings related to maintaining both muscle mass and optimal contractile function of respiratory muscles.
Collapse
Affiliation(s)
- Heather M Gransee
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | | | | |
Collapse
|
85
|
Putting the spice in weaning*. Crit Care Med 2012; 40:1022-3. [DOI: 10.1097/ccm.0b013e31823e96c2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
86
|
Wu CL, Kandarian SC. Protein overexpression in skeletal muscle using plasmid-based gene transfer to elucidate mechanisms controlling fiber size. Methods Mol Biol 2012; 798:231-243. [PMID: 22130840 DOI: 10.1007/978-1-61779-343-1_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Plasmid DNA electrotransfer is a direct method of gene delivery to skeletal muscle commonly used to identify endogenous signaling pathways that mediate muscle remodeling or pathological states in adult rodents. When plasmids encoding a protein to be overexpressed are fused to a fluorescent protein or an epitope-tag, plasmid electrotransfer permits visualization of the expressed protein in muscle fibers. Here, we demonstrate the use of electrotransfer of plasmids encoding mutant or wild type proteins to identify the role of the endogenous protein in regulating muscle fiber atrophy. The plasmids used encode a dominant negative form of the inhibitor of kappaB kinase beta (IKKβ) fused to green fluorescent protein (GFP), a constitutively active form of IKKα fused to GFP, and a wild type IKKβ fused to an HA tag. We show the effects of overexpression of these proteins on rat or mouse fiber size either with disuse atrophy or in normal weight bearing muscle. The effects of overexpressed proteins on myofiber size are assessed by comparing cross-sectional area of the transfected, fluorescent myofibers to the nontransfected, nonfluorescent myofibers. Using optimized intramuscular plasmid DNA injection and electroporation, we illustrate high transfection efficiency with no overt muscle damage using medium sized fusion proteins (105 kDa).
Collapse
Affiliation(s)
- Chia-Ling Wu
- Department of Health Sciences, Boston University, Boston, MA, USA
| | | |
Collapse
|
87
|
Abstract
Inhibition of the inhibitor of kappa B kinase (IKK)/nuclear factor-kappa B (NF-κB) pathway enhances muscle regeneration in injured and diseased skeletal muscle, but it is unclear exactly how this pathway contributes to the regeneration process. In this study, we examined the role of NF-κB in regulating the proliferation and differentiation of muscle-derived stem cells (MDSCs). MDSCs isolated from the skeletal muscles of p65(+/-) mice (haploinsufficient for the p65 subunit of NF-κB) had enhanced proliferation and myogenic differentiation compared to MDSCs isolated from wild-type (wt) littermates. In addition, selective pharmacological inhibition of IKKβ, an upstream activator of NF-κB, enhanced wt MDSC differentiation into myotubes in vitro. The p65(+/-) MDSCs also displayed a higher muscle regeneration index than wt MDSCs following implantation into adult mice with muscular dystrophy. Additionally, using a muscle injury model, we observed that p65(+/-) MDSC engraftments were associated with reduced inflammation and necrosis. These results suggest that inhibition of the IKK/NF-κB pathway represents an effective approach to improve the myogenic regenerative potential of MDSCs and possibly other adult stem cell populations. Moreover, our results suggest that the improved muscle regeneration observed following inhibition of IKK/NF-κB, is mediated, at least in part, through enhanced stem cell proliferation and myogenic potential.
Collapse
|
88
|
Braun TP, Zhu X, Szumowski M, Scott GD, Grossberg AJ, Levasseur PR, Graham K, Khan S, Damaraju S, Colmers WF, Baracos VE, Marks DL. Central nervous system inflammation induces muscle atrophy via activation of the hypothalamic-pituitary-adrenal axis. ACTA ACUST UNITED AC 2011; 208:2449-63. [PMID: 22084407 PMCID: PMC3256966 DOI: 10.1084/jem.20111020] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Systemic and CNS-delimited inflammation triggers skeletal muscle catabolism in a manner dependent on glucocorticoid signaling. Skeletal muscle catabolism is a co-morbidity of many chronic diseases and is the result of systemic inflammation. Although direct inflammatory cytokine action on muscle promotes atrophy, nonmuscle sites of action for inflammatory mediators are less well described. We demonstrate that central nervous system (CNS)–delimited interleukin 1β (IL-1β) signaling alone can evoke a catabolic program in muscle, rapidly inducing atrophy. This effect is dependent on hypothalamic–pituitary–adrenal (HPA) axis activation, as CNS IL-1β–induced atrophy is abrogated by adrenalectomy. Furthermore, we identified a glucocorticoid-responsive gene expression pattern conserved in models of acute and chronic inflammatory muscle atrophy. In contrast with studies suggesting that the direct action of inflammatory cytokines on muscle is sufficient to induce catabolism, adrenalectomy also blocks the atrophy program in response to systemic inflammation, demonstrating that glucocorticoids are requisite for this process. Additionally, circulating levels of glucocorticoids equivalent to those produced under inflammatory conditions are sufficient to cause profound muscle wasting. Together, these data suggest that a significant component of inflammation-induced muscle catabolism occurs indirectly via a relay in the CNS.
Collapse
Affiliation(s)
- Theodore P Braun
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Hutchins-Wiese HL, Li Y, Hannon K, Watkins BA. Hind limb suspension and long-chain omega-3 PUFA increase mRNA endocannabinoid system levels in skeletal muscle. J Nutr Biochem 2011; 23:986-93. [PMID: 22051448 DOI: 10.1016/j.jnutbio.2011.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 04/18/2011] [Accepted: 05/13/2011] [Indexed: 11/28/2022]
Abstract
Muscle disuse has numerous physiological consequences that end up with significant catabolic metabolism and ultimately tissue atrophy. What is not known is how muscle atrophy affects the endocannabinoid (EC) system. Arachidonic acid (AA) is the substrate for anandamide (AEA) and 2-arachidonylgycerol (2-AG), which act as agonists for cannabinoid receptors CB1 and CB2 found in muscle. Diets with n-3 polyunsaturated fatty acids (PUFA) have been shown to reduce tissue levels of AA, AEA and 2-AG. Therefore, we hypothesized that hind limb suspension (HS)-induced muscle atrophy and intake of n-3 PUFA will change mRNA levels of the EC system. Mice were randomized and assigned to a moderate n-3 PUFA [11.7 g/kg eicosapentaenoic acid (EPA)+docosahexaenoic acid (DHA)], high n-3 PUFA (17.6 g/kg EPA+DHA) or control diets for 12 days and then subjected to HS or continued weight bearing (WB) for 14 days. HS resulted in body weight, epididymal fat pad and quadriceps muscle loss compared to WB. Compared to WB, HS had greater mRNA levels of AEA and 2-AG synthesis enzymes and CB2 in the atrophied quadriceps muscle. The high n-3 PUFA diet resulted in greater mRNA levels of EC synthesis enzymes, and CB1 and CB2. The higher mRNA levels for EC with HS and dietary n-3 PUFA suggest that muscle disuse and diet induce changes in the EC system to sensitize muscle in response to metabolic and physiological consequences of atrophy.
Collapse
|
90
|
Chen KH, Cheng ML, Jing YH, Chiu DTY, Shiao MS, Chen JK. Resveratrol ameliorates metabolic disorders and muscle wasting in streptozotocin-induced diabetic rats. Am J Physiol Endocrinol Metab 2011; 301:E853-63. [PMID: 21791624 DOI: 10.1152/ajpendo.00048.2011] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus (DM) is characterized by dysregulated energy metabolism. Resveratrol (RSV) has been shown to ameliorate hyperglycemia and hyperlipidemia in diabetic animals. However, its overall in vivo effects on energy metabolism and the underlying mechanism require further investigation. In the present study, electrospray ionization-tandem mass spectrometry was employed to characterize the urine and plasma metabolomes of control, streptozotocin-induced DM and RSV-treated DM rats. Using principal component analysis (PCA) and heat map analysis, we discovered significant differences among control and experimental groups. RSV treatment significantly reduced the metabolic abnormalities in DM rats. Compared with the age-matched control rats, the level of carnitine was lower, and the levels of acetylcarnitine and butyrylcarnitine were higher in the urine and plasma of DM rats. RSV treatment ameliorated the deranged carnitine metabolism in DM rats. In addition, RSV treatment attenuated the diabetic ketoacidosis and muscle protein degradation, as evidenced from the attenuation of elevated urinary methyl-histidine and plasma branched-chain amino acids levels in DM rats. The beneficial effects of RSV in DM rats were correlated with activation of hepatic AMP-activated protein kinase and SIRT1 expression, increase of hepatic and muscular mitochondrial biogenesis and inhibition of muscle NF-κB activities. We concluded that RSV possesses multiple beneficial metabolic effects in insulin-deficient DM rats, particularly in improving energy metabolism and reducing protein wasting.
Collapse
MESH Headings
- Adenylate Kinase/genetics
- Adenylate Kinase/metabolism
- Animals
- Antioxidants/pharmacology
- Antioxidants/therapeutic use
- Cytokines/genetics
- Cytokines/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/genetics
- Drug Evaluation, Preclinical
- Male
- Metabolic Diseases/etiology
- Metabolic Diseases/genetics
- Metabolic Diseases/metabolism
- Metabolic Diseases/prevention & control
- Models, Biological
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Diseases/etiology
- Muscular Diseases/genetics
- Muscular Diseases/metabolism
- Muscular Diseases/prevention & control
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Rats
- Rats, Sprague-Dawley
- Resveratrol
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Stilbenes/pharmacology
- Stilbenes/therapeutic use
- Streptozocin
- Wasting Syndrome/etiology
- Wasting Syndrome/genetics
- Wasting Syndrome/metabolism
- Wasting Syndrome/prevention & control
Collapse
Affiliation(s)
- Kuan-Hsing Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | | | | | | | | | | |
Collapse
|
91
|
Yasuhara K, Ohno Y, Kojima A, Uehara K, Beppu M, Sugiura T, Fujimoto M, Nakai A, Ohira Y, Yoshioka T, Goto K. Absence of heat shock transcription factor 1 retards the regrowth of atrophied soleus muscle in mice. J Appl Physiol (1985) 2011; 111:1142-9. [DOI: 10.1152/japplphysiol.00471.2011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Effects of heat shock transcription factor 1 (HSF1) gene on the regrowth of atrophied mouse soleus muscles were studied. Both HSF1-null and wild-type mice were subjected to continuous hindlimb suspension for 2 wk followed by 4 wk of ambulation recovery. There was no difference in the magnitude of suspension-related decrease of muscle weight, protein content, and the cross-sectional area of muscle fibers between both types of mice. However, the regrowth of atrophied soleus muscle in HSF1-null mice was slower compared with that in wild-type mice. Lower baseline expression level of HSP25, HSC70, and HSP72 were noted in soleus muscle of HSF1-null mice. Unloading-associated downregulation and reloading-associated upregulation of HSP25 and HSP72 mRNA were observed not only in wild-type mice but also in HSF1-null mice. Reloading-associated upregulation of HSP72 and HSP25 during the regrowth of atrophied muscle was observed in wild-type mice. Minor and delayed upregulation of HSP72 at mRNA and protein levels was also seen in HSF1-null mice. Significant upregulations of HSF2 and HSF4 were observed immediately after the suspension in HSF1-null mice, but not in wild-type mice. Therefore, HSP72 expression in soleus muscle might be regulated by the posttranscriptional level, but not by the stress response. Evidence from this study suggested that the upregulation of HSPs induced by HSF1-associated stress response might play, in part, important roles in the mechanical loading (stress)-associated regrowth of skeletal muscle.
Collapse
Affiliation(s)
- Kazuyuki Yasuhara
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Kawasaki
| | - Yoshitaka Ohno
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University, Toyohashi
| | - Atsushi Kojima
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Kawasaki
| | - Kenji Uehara
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Kawasaki
| | - Moroe Beppu
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Kawasaki
| | | | | | - Akira Nakai
- Graduate School of Medicine, Yamaguchi University, Yamaguchi
| | | | | | - Katsumasa Goto
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Toyohashi, Japan
| |
Collapse
|
92
|
Caron AZ, Haroun S, Leblanc E, Trensz F, Guindi C, Amrani A, Grenier G. The proteasome inhibitor MG132 reduces immobilization-induced skeletal muscle atrophy in mice. BMC Musculoskelet Disord 2011; 12:185. [PMID: 21843349 PMCID: PMC3173404 DOI: 10.1186/1471-2474-12-185] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 08/15/2011] [Indexed: 11/10/2022] Open
Abstract
Background Skeletal muscle atrophy is a serious concern for the rehabilitation of patients afflicted by prolonged limb restriction. This debilitating condition is associated with a marked activation of NFκB activity. The ubiquitin-proteasome pathway degrades the NFκB inhibitor IκBα, enabling NFκB to translocate to the nucleus and bind to the target genes that promote muscle atrophy. Although several studies showed that proteasome inhibitors are efficient to reduce atrophy, no studies have demonstrated the ability of these inhibitors to preserve muscle function under catabolic condition. Methods We recently developed a new hindlimb immobilization procedure that induces significant skeletal muscle atrophy and used it to show that an inflammatory process characterized by the up-regulation of TNFα, a known activator of the canonical NFκB pathway, is associated with the atrophy. Here, we used this model to investigate the effect of in vivo proteasome inhibition on the muscle integrity by histological approach. TNFα, IL-1, IL-6, MuRF-1 and Atrogin/MAFbx mRNA level were determined by qPCR. Also, a functional measurement of locomotors activity was performed to determine if the treatment can shorten the rehabilitation period following immobilization. Results In the present study, we showed that the proteasome inhibitor MG132 significantly inhibited IκBα degradation thus preventing NFκB activation in vitro. MG132 preserved muscle and myofiber cross-sectional area by downregulating the muscle-specific ubiquitin ligases atrogin-1/MAFbx and MuRF-1 mRNA in vivo. This effect resulted in a diminished rehabilitation period. Conclusion These finding demonstrate that proteasome inhibitors show potential for the development of pharmacological therapies to prevent muscle atrophy and thus favor muscle rehabilitation.
Collapse
Affiliation(s)
- Annabelle Z Caron
- Centre de Recherche Clinique Étienne-Lebel, 3001-12th Avenue North, Sherbrooke, QC J1H5N4, Canada
| | | | | | | | | | | | | |
Collapse
|
93
|
Wing SS, Lecker SH, Jagoe RT. Proteolysis in illness-associated skeletal muscle atrophy: from pathways to networks. Crit Rev Clin Lab Sci 2011; 48:49-70. [PMID: 21699435 DOI: 10.3109/10408363.2011.586171] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Improvements in health in the past decades have resulted in increased numbers of the elderly in both developed and developing regions of the world. Advances in therapy have also increased the prevalence of patients with chronic and degenerative diseases. Muscle wasting, a feature of most chronic diseases, is prominent in the elderly and contributes to both morbidity and mortality. A major research goal has been to identify the proteolytic system(s) that is responsible for the degradation of proteins that occurs in muscle atrophy. Findings over the past 20 years have clearly confirmed an important role of the ubiquitin proteasome system in mediating muscle proteolysis, particularly that of myofibrillar proteins. However, recent observations have provided evidence that autophagy, calpains and caspases also contribute to the turnover of muscle proteins in catabolic states, and furthermore, that these diverse proteolytic systems interact with each other at various levels. Importantly, a number of intracellular signaling pathways such as the IGF1/AKT, myostatin/Smad, PGC1, cytokine/NFκB, and AMPK pathways are now known to interact and can regulate some of these proteolytic systems in a coordinated manner. A number of loss of function studies have identified promising therapeutic approaches to the prevention and treatment of wasting. However, additional biomarkers and other approaches to improve early identification of patients who would benefit from such treatment need to be developed. The current data suggests a network of interacting proteolytic and signaling pathways in muscle. Future studies are needed to improve understanding of the nature and control of these interactions and how they work to preserve muscle function under various states of growth and atrophy.
Collapse
Affiliation(s)
- Simon S Wing
- Departments of Medicine, McGill University and McGill University Health Centre Research Institute, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
94
|
Shadfar S, Couch ME, McKinney KA, Weinstein LJ, Yin X, Rodríguez JE, Guttridge DC, Willis M. Oral resveratrol therapy inhibits cancer-induced skeletal muscle and cardiac atrophy in vivo. Nutr Cancer 2011; 63:749-62. [PMID: 21660860 DOI: 10.1080/01635581.2011.563032] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The mechanism by which cancer mediates muscle atrophy has been delineated in the past 3 decades and includes a prominent role of tumor-derived cytokines, such as IL-6, TNFα, and IL-1. These cytokines interact with their cognate receptors on muscle to activate the downstream transcription factor NF-κB and induce sarcomere proteolysis. Experimentally, inhibiting NF-κB signaling largely prevents cancer-induced muscle wasting, indicating its prominent role in muscle atrophy. Resveratrol, a natural phytoalexin found in the skin of grapes, has recently been shown to inhibit NF-κB in cancer cells, which led us to hypothesize that it might have a protective role in cancer cachexia. Therefore, we investigated whether daily oral resveratrol could protect against skeletal muscle loss and cardiac atrophy in an established mouse model. We demonstrate resveratrol inhibits skeletal muscle and cardiac atrophy induced by C26 adenocarcinoma tumors through its inhibition of NF-κB (p65) activity in skeletal muscle and heart. These studies demonstrate for the first time the utility of oral resveratrol therapy to provide clinical benefit in cancer-induced atrophy through the inhibition of NF-κB in muscle. These findings may have application in the treatment of diseases with parallel pathophysiologies such as muscular dystrophy and heart failure.
Collapse
Affiliation(s)
- Scott Shadfar
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Kossler N, Stricker S, Rödelsperger C, Robinson PN, Kim J, Dietrich C, Osswald M, Kühnisch J, Stevenson DA, Braun T, Mundlos S, Kolanczyk M. Neurofibromin (Nf1) is required for skeletal muscle development. Hum Mol Genet 2011; 20:2697-709. [PMID: 21478499 DOI: 10.1093/hmg/ddr149] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) is a multi-system disease caused by mutations in the NF1 gene encoding a Ras-GAP protein, neurofibromin, which negatively regulates Ras signaling. Besides neuroectodermal malformations and tumors, the skeletal system is often affected (e.g. scoliosis and long bone dysplasia) demonstrating the importance of neurofibromin for development and maintenance of the musculoskeletal system. Here, we focus on the role of neurofibromin in skeletal muscle development. Nf1 gene inactivation in the early limb bud mesenchyme using Prx1-cre (Nf1(Prx1)) resulted in muscle dystrophy characterized by fibrosis, reduced number of muscle fibers and reduced muscle force. This was caused by an early defect in myogenesis affecting the terminal differentiation of myoblasts between E12.5 and E14.5. In parallel, the muscle connective tissue cells exhibited increased proliferation at E14.5 and an increase in the amount of connective tissue as early as E16.5. These changes were accompanied by excessive mitogen-activated protein kinase pathway activation. Satellite cells isolated from Nf1(Prx1) mice showed normal self-renewal, but their differentiation was impaired as indicated by diminished myotube formation. Our results demonstrate a requirement of neurofibromin for muscle formation and maintenance. This previously unrecognized function of neurofibromin may contribute to the musculoskeletal problems in NF1 patients.
Collapse
Affiliation(s)
- Nadine Kossler
- FG Development & Disease, Max Planck Institute for Molecular Genetics, Ihnestrasse 73, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Liu X, Manzano G, Kim HT, Feeley BT. A rat model of massive rotator cuff tears. J Orthop Res 2011; 29:588-95. [PMID: 20949443 DOI: 10.1002/jor.21266] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 08/23/2010] [Indexed: 02/04/2023]
Abstract
Rotator cuff tears (RCTs) are the most common tendon injury seen in orthopedic patients. Massive RCT does not heal spontaneously and results in poor clinical outcomes. Muscle atrophy and fatty infiltration in rotator cuff muscles are major complications of chronic massive RCT and are thought to be the key factors responsible for the failure of attempted massive RCT repair. However, the pathophysiology of rotator cuff muscle atrophy and fat infiltration remains largely unknown, and no small animal model has been shown to reproduce the histologic and molecular changes seen in massive RCT. In this article, we report a novel rat massive RCT model, in which significant and consistent muscle atrophy and fat infiltration were observed in the rotator cuff muscles after rotator cuff tendon transection and denervation. The supraspinatus and infraspinatus muscle lost 25.4% and 28.9% of their wet weight 2 weeks after complete tendon transection, respectively. Six weeks after surgery, the average wet weight of supraspinatus and infraspinatus muscles decreased 13.2% and 28.3%, respectively. Significant fat infiltration was only observed in infraspinatus 6 weeks after tendon transection.
Collapse
Affiliation(s)
- Xuhui Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | | | | | | |
Collapse
|
97
|
Dagdeviren S, Kandilci HB, Uysal B, Zeybek ND, Korkusuz P, Gümüsel B, Korkusuz F. Tumor necrosis factor-alpha antagonist administration recovers skeletal muscle dysfunction in ovariectomized rats. J Orthop Res 2011; 29:275-80. [PMID: 20690186 DOI: 10.1002/jor.21226] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Skeletal muscles deteriorate after ovariectomy. Molecular pathway of this deterioration has not been defined. Tumor necrosis factor (TNF)-alpha activation is assumed to trigger muscle atrophy and administration of its antagonist is hypothesized to recover this atrophy in rats. Slow-twitch soleus and fast-twitch extensor digitorum longus muscle functions were investigated in intact, ovariectomized (OVX), and OVX plus 10 µg/g/week TNF-alpha antagonist administered female rats. Maximum isometric twitch and tetanic contraction responses were lower in the OVX groups. Maximum isometric twitch amplitudes recovered in the extensor digitorum longus but not in the soleus muscles after TNF-alpha antagonist administration. The decrease in responses to tetanic stimulations recovered in the OVX-TNF group at frequencies higher than 20 Hz in both muscle types. OVX animals body weight was 21% higher than intact animals. Muscle weight to body weight ratios of the OVX groups were higher than the control group which recovered after TNF-alpha antagonist administration. Findings suggest that the functional loss in OVX rat muscles is TNF-alpha pathway dependent. Skeletal muscle atrophy and function after OVX recovered by TNF-alpha antagonist administration.
Collapse
Affiliation(s)
- Sezin Dagdeviren
- Medical Center, Department of Biotechnology, Middle East Technical University, İnönü Bulvarı, Ankara 06531, Turkey
| | | | | | | | | | | | | |
Collapse
|
98
|
Reed SA, Senf SM, Cornwell EW, Kandarian SC, Judge AR. Inhibition of IkappaB kinase alpha (IKKα) or IKKbeta (IKKβ) plus forkhead box O (Foxo) abolishes skeletal muscle atrophy. Biochem Biophys Res Commun 2011; 405:491-6. [PMID: 21256828 DOI: 10.1016/j.bbrc.2011.01.059] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/17/2011] [Indexed: 10/18/2022]
Abstract
Two transcription factor families that are activated during multiple conditions of skeletal muscle wasting are nuclear factor κB (NF-κB) and forkhead box O (Foxo). There is clear evidence that both NF-κB and Foxo activation are sufficient to cause muscle fiber atrophy and they are individually required for at least half of the fiber atrophy during muscle disuse, but there is no work determining the combined effect of inhibiting these factors during a physiological condition of muscle atrophy. Here, we determined whether inhibition of Foxo activation plus inhibition of NF-κB activation, the latter by blocking the upstream inhibitor of kappaB kinases (IKKα and IKKβ), would prevent muscle atrophy induced by 7 days of cast immobilization. Results were based on measurements of mean fiber cross-sectional area (CSA) from 72 muscles transfected with 5 different mutant expression plasmids or plasmid combinations. Immobilization caused a 47% decrease in fiber CSA in muscles injected with control plasmids. Fibers from immobilized muscles transfected with dominant negative (d.n.) IKKα-EGFP, d.n. IKKβ-EGFP or d.n. Foxo-DsRed showed a 22%, 57%, and 76% inhibition of atrophy, respectively. Co-expression of d.n. IKKα-EGFP and d.n. Foxo-DsRed significantly inhibited 89% of the immobilization-induced fiber atrophy. Similarly, co-expression of d.n. IKKβ-EGFP and d.n. Foxo-DsRed inhibited the immobilization-induced fiber atrophy by 95%. These findings demonstrate that the combined effects of inhibiting immobilization-induced NF-κB and Foxo transcriptional activity has an additive effect on preventing immobilization-induced atrophy, indicating that NF-κB and Foxo have a cumulative effect on atrophy signaling and/or atrophy gene expression.
Collapse
Affiliation(s)
- S A Reed
- Department of Physical Therapy, University of Florida, 101 S. Newell Drive, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
99
|
Wu CL, Kandarian SC, Jackman RW. Identification of genes that elicit disuse muscle atrophy via the transcription factors p50 and Bcl-3. PLoS One 2011; 6:e16171. [PMID: 21249144 PMCID: PMC3020958 DOI: 10.1371/journal.pone.0016171] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 12/09/2010] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle atrophy is a debilitating condition associated with weakness, fatigue, and reduced functional capacity. Nuclear factor-kappaB (NF-κB) transcription factors play a critical role in atrophy. Knockout of genes encoding p50 or the NF-κB co-transactivator, Bcl-3, abolish disuse atrophy and thus they are NF-κB factors required for disuse atrophy. We do not know however, the genes targeted by NF-κB that produce the atrophied phenotype. Here we identify the genes required to produce disuse atrophy using gene expression profiling in wild type compared to Nfkb1 (gene encodes p50) and Bcl-3 deficient mice. There were 185 and 240 genes upregulated in wild type mice due to unloading, that were not upregulated in Nfkb1−/− and Bcl-3−/− mice, respectively, and so these genes were considered direct or indirect targets of p50 and Bcl-3. All of the p50 gene targets were contained in the Bcl-3 gene target list. Most genes were involved with protein degradation, signaling, translation, transcription, and transport. To identify direct targets of p50 and Bcl-3 we performed chromatin immunoprecipitation of selected genes previously shown to have roles in atrophy. Trim63 (MuRF1), Fbxo32 (MAFbx), Ubc, Ctsl, Runx1, Tnfrsf12a (Tweak receptor), and Cxcl10 (IP-10) showed increased Bcl-3 binding to κB sites in unloaded muscle and thus were direct targets of Bcl-3. p50 binding to the same sites on these genes either did not change or increased, supporting the idea of p50:Bcl-3 binding complexes. p65 binding to κB sites showed decreased or no binding to these genes with unloading. Fbxo9, Psma6, Psmc4, Psmg4, Foxo3, Ankrd1 (CARP), and Eif4ebp1 did not show changes in p65, p50, or Bcl-3 binding to κB sites, and so were considered indirect targets of p50 and Bcl-3. This work represents the first study to use a global approach to identify genes required to produce the atrophied phenotype with disuse.
Collapse
Affiliation(s)
- Chia-Ling Wu
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Susan C. Kandarian
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Robert W. Jackman
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
100
|
Hain BA, Dodd SL, Judge AR. IκBα degradation is necessary for skeletal muscle atrophy associated with contractile claudication. Am J Physiol Regul Integr Comp Physiol 2011; 300:R595-604. [PMID: 21209383 DOI: 10.1152/ajpregu.00728.2010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The arterial blockage in patients with peripheral arterial disease (PAD) restricts oxygen delivery to skeletal muscles distal to the blockage. In advanced-stage PAD patients, this creates a chronic ischemic condition in the affected muscles. However, in the majority of PAD patients, the muscles distal to the blockage only become ischemic during physical activity when the oxygen demands of these muscles are increased. Therefore, the skeletal muscle of most PAD patients undergoes repeated cycles of low-grade ischemia-reperfusion each time the patient is active and then rests. This has been speculated to contribute to the biochemical and morphological myopathies observed in PAD patients. The current study aimed to determine, using a rodent model, whether repeated hind limb muscle contractions during blood flow restriction to the hind limb muscles increases NF-κB activity. We, subsequently, determined whether an increase in NF-κB activity during this condition is required for the increased transcription of specific atrophy-related genes and muscle fiber atrophy. We found that hind limb muscle contractions during blood flow restriction to the limb increased NF-κB activity, the transcription of specific atrophy-related genes, and caused a 35% decrease in muscle fiber cross-sectional area. We further found that inhibition of NF-κB activity, via gene transfer of a dominant-negative inhibitor of κBα (d.n. IκBα), prevented the increase in atrophy gene expression and muscle fiber atrophy. These findings demonstrate that when blood flow to skeletal muscle is restricted, repeated cycles of muscle contraction can cause muscle fiber atrophy that requires NF-κB-IκBα signaling.
Collapse
Affiliation(s)
- Brian A Hain
- Department of Applied Physiology, Univ. of Florida, Gainesville, 32611, USA
| | | | | |
Collapse
|