Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Oncotarget ; 7(33): 52695-52709, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27447862

ABSTRACT

Fatigue is a symptom of many diseases, but it can also manifest as a unique medical condition, such as idiopathic chronic fatigue (ICF). While the prevalence of ICF increases with age, mitochondrial content and function decline with age, which may contribute to ICF. The purpose of this study was to determine whether skeletal muscle mitochondrial dysregulation and oxidative stress is linked to ICF in older adults. Sedentary, old adults (n = 48, age 72.4 ± 5.3 years) were categorized into ICF and non-fatigued (NF) groups based on the FACIT-Fatigue questionnaire. ICF individuals had a FACIT score one standard deviation below the mean for non-anemic adults > 65 years and were excluded according to CDC diagnostic criteria for ICF. Vastus lateralis muscle biopsies were analyzed, showing reductions in mitochondrial content and suppression of mitochondrial regulatory proteins Sirt3, PGC-1α, NRF-1, and cytochrome c in ICF compared to NF. Additionally, mitochondrial morphology proteins, antioxidant enzymes, and lipid peroxidation were unchanged in ICF individuals. Our data suggests older adults with ICF have reduced skeletal muscle mitochondrial content and biogenesis signaling that cannot be accounted for by increased oxidative damage.


Subject(s)
Fatigue Syndrome, Chronic/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Signal Transduction , Aged , Antioxidants/metabolism , Cytochromes c/metabolism , Fatigue/diagnosis , Fatigue/etiology , Fatigue/metabolism , Fatigue Syndrome, Chronic/diagnosis , Fatigue Syndrome, Chronic/etiology , Female , Humans , Male , NF-E2-Related Factor 1/metabolism , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 3/metabolism , Surveys and Questionnaires
2.
J Appl Physiol (1985) ; 120(7): 733-42, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26846552

ABSTRACT

Diaphragm muscle weakness in chronic heart failure (CHF) is caused by elevated oxidants and exacerbates breathing abnormalities, exercise intolerance, and dyspnea. However, the specific source of oxidants that cause diaphragm weakness is unknown. We examined whether mitochondrial reactive oxygen species (ROS) cause diaphragm weakness in CHF by testing the hypothesis that CHF animals treated with a mitochondria-targeted antioxidant have normal diaphragm function. Rats underwent CHF or sham surgery. Eight weeks after surgeries, we administered a mitochondrial-targeted antioxidant (MitoTEMPO; 1 mg·kg(-1)·day(-1)) or sterile saline (Vehicle). Left ventricular dysfunction (echocardiography) pre- and posttreatment and morphological abnormalities were consistent with the presence of CHF. CHF elicited a threefold (P < 0.05) increase in diaphragm mitochondrial H2O2 emission, decreased diaphragm glutathione content by 23%, and also depressed twitch and maximal tetanic force by ∼20% in Vehicle-treated animals compared with Sham (P < 0.05 for all comparisons). Diaphragm mitochondrial H2O2 emission, glutathione content, and twitch and maximal tetanic force were normal in CHF animals receiving MitoTEMPO. Neither CHF nor MitoTEMPO altered the diaphragm protein levels of antioxidant enzymes: superoxide dismutases (CuZn-SOD or MnSOD), glutathione peroxidase, and catalase. In both Vehicle and MitoTEMPO groups, CHF elicited a ∼30% increase in cytochrome c oxidase activity, whereas there were no changes in citrate synthase activity. Our data suggest that elevated mitochondrial H2O2 emission causes diaphragm weakness in CHF. Moreover, changes in protein levels of antioxidant enzymes or mitochondrial content do not seem to mediate the increase in mitochondria H2O2 emission in CHF and protective effects of MitoTEMPO.


Subject(s)
Diaphragm/physiopathology , Heart Failure/physiopathology , Mitochondria/metabolism , Muscle Weakness/physiopathology , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Diaphragm/metabolism , Electron Transport Complex IV/metabolism , Glutathione/metabolism , Glutathione Peroxidase , Heart Failure/metabolism , Hydrogen Peroxide/metabolism , Muscle Weakness/metabolism , Oxidation-Reduction , Oxidative Stress/physiology , Rats , Rats, Wistar , Superoxides/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
4.
J Physiol ; 594(18): 5105-23, 2016 09 15.
Article in English | MEDLINE | ID: mdl-26503074

ABSTRACT

Mitochondria are negatively affected by ageing leading to their inability to adapt to higher levels of oxidative stress and this ultimately contributes to the systemic loss of muscle mass and function termed sarcopenia. Since mitochondria are central mediators of muscle health, they have become highly sought-after targets of physiological and pharmacological interventions. Exercise is the only known strategy to combat sarcopenia and this is largely mediated through improvements in mitochondrial plasticity. More recently a critical role for mitochondrial turnover in preserving muscle has been postulated. Specifically, cellular pathways responsible for the regulation of mitochondrial turnover including biogenesis, dynamics and autophagy may become dysregulated during ageing resulting in the reduced clearance and accumulation of damaged organelles within the cell. When mitochondrial quality is compromised and homeostasis is not re-established, myonuclear cell death is activated and muscle atrophy ensues. In contrast, acute and chronic exercise attenuates these deficits, restoring mitochondrial turnover and promoting a healthier mitochondrial pool that leads to the preservation of muscle. Additionally, the magnitude of these exercise-induced mitochondrial adaptations is currently debated with several studies reporting a lower adaptability of old muscle relative to young, but the processes responsible for this diminished training response are unclear. Based on these observations, understanding the molecular details of how advancing age and exercise influence mitochondria in older muscle will provide invaluable insight into the development of exercise protocols that will maximize beneficial adaptations in the elderly. This information will also be imperative for future research exploring pharmacological targets of mitochondrial plasticity.


Subject(s)
Aging/physiology , Exercise/physiology , Mitochondria, Muscle/physiology , Muscle, Skeletal/physiology , Oxidative Stress/physiology , Animals , Humans , Muscular Atrophy/physiopathology
6.
Mitochondrion ; 14(1): 42-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24246911

ABSTRACT

Near-infrared (NIR) light is a complementary therapy used to treat musculoskeletal injuries but the underlying mechanisms are unclear. Acute NIR light treatment (~800-950 nm; 22.8 J/cm(2)) induced a dose-dependent increase in mitochondrial signaling (AMPK, p38 MAPK) in differentiated muscle cells. Repeated NIR light exposure (4 days) appeared to elevate oxidative stress and increase the upstream mitochondrial regulatory proteins AMPK (3.1-fold), p38 (2.8-fold), PGC-1α (19.7%), Sirt1 (26.8%), and reduced RIP140 (23.2%), but downstream mitochondrial regulation/content (Tfam, NRF-1, Sirt3, cytochrome c, ETC subunits) was unaltered. Our data indicates that NIR light alters mitochondrial biogenesis signaling and may represent a mechanistic link to the clinical benefits.


Subject(s)
Infrared Rays , Mitochondria/physiology , Mitochondria/radiation effects , Mitochondrial Turnover/radiation effects , Muscle Cells/physiology , Muscle Cells/radiation effects , Signal Transduction , Animals , Mice
7.
PLoS One ; 8(7): e69327, 2013.
Article in English | MEDLINE | ID: mdl-23935986

ABSTRACT

Mitochondrial DNA (mtDNA) mutations lead to decrements in mitochondrial function and accelerated rates of these mutations has been linked to skeletal muscle loss (sarcopenia). The purpose of this study was to investigate the effect of mtDNA mutations on mitochondrial quality control processes in skeletal muscle from animals (young; 3-6 months and older; 8-15 months) expressing a proofreading-deficient version of mtDNA polymerase gamma (PolG). This progeroid aging model exhibits elevated mtDNA mutation rates, mitochondrial dysfunction, and a premature aging phenotype that includes sarcopenia. We found increased expression of the mitochondrial biogenesis regulator peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and its target proteins, nuclear respiratory factor 1 (NRF-1) and mitochondrial transcription factor A (Tfam) in PolG animals compared to wild-type (WT) (P<0.05). Muscle from older PolG animals displayed higher mitochondrial fission protein 1 (Fis1) concurrent with greater induction of autophagy, as indicated by changes in Atg5 and p62 protein content (P<0.05). Additionally, levels of the Tom22 import protein were higher in PolG animals when compared to WT (P<0.05). In contrast, muscle from normally-aged animals exhibited a distinctly different expression profile compared to PolG animals. Older WT animals appeared to have higher fusion (greater Mfn1/Mfn2, and lower Fis1) and lower autophagy (Beclin-1 and p62) compared to young WT suggesting that autophagy is impaired in aging muscle. In conclusion, muscle from mtDNA mutator mice display higher mitochondrial fission and autophagy levels that likely contribute to the sarcopenic phenotype observed in premature aging and this differs from the response observed in normally-aged muscle.


Subject(s)
Aging, Premature/pathology , Mitochondria/metabolism , Sarcopenia/pathology , Aging, Premature/metabolism , Animals , Autophagy , DNA Polymerase gamma , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/metabolism , Disease Models, Animal , Mice , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Muscles/enzymology , Muscles/pathology , Protein Transport , Sarcopenia/metabolism , Up-Regulation
8.
Exp Gerontol ; 48(9): 858-68, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23747682

ABSTRACT

Aging is associated with a loss in muscle known as sarcopenia that is partially attributed to apoptosis. In aging rodents, caloric restriction (CR) increases health and longevity by improving mitochondrial function and the polyphenol resveratrol (RSV) has been reported to have similar benefits. In the present study, we investigated the potential efficacy of using short-term (6 weeks) CR (20%), RSV (50 mg/kg/day), or combined CR+ RSV (20% CR and 50 mg/kg/day RSV), initiated at late-life (27 months) to protect muscle against sarcopenia by altering mitochondrial function, biogenesis, content, and apoptotic signaling in both glycolytic white and oxidative red gastrocnemius muscle (WG and RG, respectively) of male Fischer 344 × Brown Norway rats. CR but not RSV attenuated the age-associated loss of muscle mass in both mixed gastrocnemius and soleus muscle, while combined treatment (CR + RSV) paradigms showed a protective effect in the soleus and plantaris muscle (P < 0.05). Sirt1 protein content was increased by 2.6-fold (P < 0.05) in WG but not RG muscle with RSV treatment, while CR or CR + RSV had no effect. PGC-1α levels were higher (2-fold) in the WG from CR-treated animals (P < 0.05) when compared to ad-libitum (AL) animals but no differences were observed in the RG with any treatment. Levels of the anti-apoptotic protein Bcl-2 were significantly higher (1.6-fold) in the WG muscle of RSV and CR + RSV groups compared to AL (P < 0.05) but tended to occur coincident with elevations in the pro-apoptotic protein Bax so that the apoptotic susceptibility as indicated by the Bax to Bcl-2 ratio was unchanged. There were no alterations in DNA fragmentation with any treatment in muscle from older animals. Additionally, mitochondrial respiration measured in permeabilized muscle fibers was unchanged in any treatment group and this paralleled the lack of change in cytochrome c oxidase (COX) activity. These data suggest that short-term moderate CR, RSV, or CR + RSV tended to modestly alter key mitochondrial regulatory and apoptotic signaling pathways in glycolytic muscle and this might contribute to the moderate protective effects against aging-induced muscle loss observed in this study.


Subject(s)
Aging/metabolism , Caloric Restriction , Mitochondrial Proteins/metabolism , Sarcopenia/prevention & control , Stilbenes/therapeutic use , AMP-Activated Protein Kinases/metabolism , Aging/drug effects , Aging/pathology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Combined Modality Therapy , Male , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Organ Size/drug effects , Oxygen Consumption/physiology , Rats , Rats, Inbred F344 , Resveratrol , Sarcopenia/metabolism , Sarcopenia/pathology , Sirtuin 1/metabolism , Stilbenes/pharmacology
9.
Biol Chem ; 394(3): 393-414, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23154422

ABSTRACT

Muscle loss during aging and disuse is a highly prevalent and disabling condition, but knowledge about cellular pathways mediating muscle atrophy is still limited. Given the postmitotic nature of skeletal myocytes, the maintenance of cellular homeostasis relies on the efficiency of cellular quality control mechanisms. In this scenario, alterations in mitochondrial function are considered a major factor underlying sarcopenia and muscle atrophy. Damaged mitochondria are not only less bioenergetically efficient, but also generate increased amounts of reactive oxygen species, interfere with cellular quality control mechanisms, and display a greater propensity to trigger apoptosis. Thus, mitochondria stand at the crossroad of signaling pathways that regulate skeletal myocyte function and viability. Studies on these pathways have sometimes provided unexpected and counterintuitive results, which suggests that they are organized into a complex, heterarchical network that is currently insufficiently understood. Untangling the complexity of such a network will likely provide clinicians with novel and highly effective therapeutics to counter the muscle loss associated with aging and disuse. In this review, we summarize the current knowledge on the mechanisms whereby mitochondrial dysfunction intervenes in the pathogenesis of sarcopenia and disuse atrophy, and highlight the prospect of targeting specific processes to treat these conditions.


Subject(s)
Mitochondria/pathology , Muscular Disorders, Atrophic/physiopathology , Sarcopenia/physiopathology , Humans , Oxidative Stress , Signal Transduction
10.
Aging Cell ; 11(5): 801-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22681576

ABSTRACT

Age-related loss of muscle mass and strength (sarcopenia) leads to a decline in physical function and frailty in the elderly. Among the many proposed underlying causes of sarcopenia, mitochondrial dysfunction is inherent in a variety of aged tissues. The intent of this study was to examine the effect of aging on key groups of regulatory proteins involved in mitochondrial biogenesis and how this relates to physical performance in two groups of sedentary elderly participants, classified as high- and low-functioning based on the Short Physical Performance Battery test. Muscle mass was decreased by 38% and 30% in low-functioning elderly (LFE) participants when compared to young and high-functioning elderly participants, respectively, and positively correlated to physical performance. Mitochondrial respiration in permeabilized muscle fibers was reduced (41%) in the LFE group when compared to the young, and this was associated with a 30% decline in cytochrome c oxidase activity. Levels of key metabolic regulators, SIRT3 and PGC-1α, were significantly reduced (50%) in both groups of elderly participants when compared to young. Similarly, the fusion protein OPA1 was lower in muscle from elderly subjects; however, no changes were detected in Mfn2, Drp1 or Fis1 among the groups. In contrast, protein import machinery components Tom22 and cHsp70 were increased in the LFE group when compared to the young. This study suggests that aging in skeletal muscle is associated with impaired mitochondrial function and altered biogenesis pathways and that this may contribute to muscle atrophy and the decline in muscle performance observed in the elderly population.


Subject(s)
Aging/physiology , Mitochondria, Muscle/physiology , Muscle, Skeletal/physiology , Sedentary Behavior , AMP-Activated Protein Kinases/metabolism , Adult , Aged , Aged, 80 and over , Aging/metabolism , Female , Humans , MAP Kinase Signaling System , Male , Middle Aged , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Sarcopenia/metabolism , Sarcopenia/pathology , Young Adult
11.
J Physiol ; 589(Pt 9): 2129-38, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21224240

ABSTRACT

It is well established that contracting skeletal muscles produce free radicals. Given that radicals are known to play a prominent role in the pathogenesis of several diseases, the 1980s-90s dogma was that contraction-induced radical production was detrimental to muscle because of oxidative damage to macromolecules within the fibre. In contrast to this early outlook, it is now clear that both reactive oxygen species (ROS) and reactive nitrogen species (RNS) play important roles in cell signalling pathways involved in muscle adaptation to exercise and the remodelling that occurs in skeletal muscle during periods of prolonged inactivity. This review will highlight two important redox sensitive signalling pathways that contribute to ROS and RNS-induced skeletal muscle adaptation to endurance exercise. We begin with a historical overview of radical production in skeletal muscles followed by a discussion of the intracellular sites for ROS and RNS production in muscle fibres. We will then provide a synopsis of the redox-sensitive NF-B and PGC-1α signalling pathways that contribute to skeletal muscle adaptation in response to exercise training. We will conclude with a discussion of unanswered questions in redox signalling in skeletal muscle in the hope of promoting additional research interest in this field.


Subject(s)
Muscle Contraction , Muscle, Skeletal/metabolism , Oxidative Stress , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Adaptation, Physiological , Animals , Heat-Shock Proteins/metabolism , Humans , Muscle, Skeletal/pathology , NF-kappa B/metabolism , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Endurance , Transcription Factors/metabolism
12.
Am J Physiol Cell Physiol ; 298(6): C1308-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20107041

ABSTRACT

Evidence exists that mitochondrial content and/or function is reduced in muscle of aging individuals. The purposes of this study were to investigate the contribution of outer membrane protein import and assembly processes to this decline and to determine whether the assembly process could adapt to chronic contractile activity (CCA). Tom40 assembly into the translocases of the outer membrane (TOM complex) was measured in subsarcolemmal mitochondria obtained from young (6 mo old) and aged (36 mo old) Fischer 344 x Brown Norway animals. While the initial import of Tom40 did not differ between young and aged animals, its subsequent assembly into the final approximately 380 kDa complex was 2.2-fold higher (P < 0.05) in mitochondria from aged compared with young animals. This was associated with a higher abundance of Tom22, a protein vital for the assembly process. CCA induced a greater initial import and subsequent assembly of Tom40 in mitochondria from young animals, resulting in a CCA-induced 75% increase (P < 0.05) in Tom40 within mitochondria. This effect of CCA was attenuated in mitochondria from old animals. These data suggest that the import and assembly of proteins into the outer membrane do not contribute to reduced mitochondrial content or function in aged animals. Indeed, the greater assembly rate in mitochondria from aged animals may be a compensatory mechanism attempting to offset any decrements in mitochondrial content or function within aged muscle. Our data also indicate the potential of CCA to contribute to increased mitochondrial biogenesis in muscle through changes in the outer membrane import and assembly pathway.


Subject(s)
Aging/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Muscle Contraction , Muscle, Skeletal/metabolism , Adaptation, Physiological , Age Factors , Animals , Crosses, Genetic , Male , Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Precursor Protein Import Complex Proteins , Protein Multimerization , Protein Transport , Rats , Rats, Inbred BN , Rats, Inbred F344
13.
J Appl Physiol (1985) ; 107(6): 1730-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19797682

ABSTRACT

Sirt1 is a NAD(+)-dependent histone deacetylase that interacts with the regulatory protein of mitochondrial biogenesis PGC-1alpha and is sensitive to metabolic alterations. We assessed whether a strict relationship between the expression of Sirt1, mitochondrial proteins, and PGC-1alpha existed across tissues possessing a wide range of oxidative capabilities, as well as in skeletal muscle subject to chronic use (voluntary wheel running or electrical stimulation for 7 days, 10 Hz; 3 h/day) or disuse (denervation for up to 21 days) in which organelle biogenesis is altered. PGC-1alpha levels were not closely associated with the expression of Sirt1, measured using immunoblotting or via enzymatic deacetylase activity. The mitochondrial protein cytochrome c increased by 70-90% in soleus and plantaris muscles of running animals, whereas Sirt1 activity remained unchanged. In chronically stimulated muscle, cytochrome c was increased by 30% compared with nonstimulated muscle, whereas Sirt1 activity was increased modestly by 20-25%. In contrast, in denervated muscle, these markers of mitochondrial content were decreased by 30-50% compared with the control muscle, whereas Sirt1 activity was increased by 75-80%. Our data suggest that Sirt1 and PGC-1alpha expression are independently regulated and that, although Sirt1 activity may be involved in mitochondrial biogenesis, its expression is not closely correlated to changes in mitochondrial proteins during conditions of chronic muscle use and disuse.


Subject(s)
Mitochondrial Proteins/metabolism , Motor Activity/physiology , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Sirtuin 1/metabolism , Adipose Tissue, White/metabolism , Animals , Blotting, Western , Cytochromes c/metabolism , Denervation , Electric Stimulation , Enzyme Assays , Liver/metabolism , Male , Mitochondria/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/innervation , Myocardium/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Transcription Factors/metabolism
14.
Am J Physiol Cell Physiol ; 297(1): C217-25, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19439529

ABSTRACT

Mitochondria are critical for cellular bioenergetics, and they mediate apoptosis within cells. We used whole body peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha) knockout (KO) animals to investigate its role on organelle function, apoptotic signaling, and cytochrome-c oxidase activity, an indicator of mitochondrial content, in muscle and other tissues (brain, liver, and pancreas). Lack of PGC-1alpha reduced mitochondrial content in all muscles (17-44%; P < 0.05) but had no effect in brain, liver, and pancreas. However, the tissue expression of proteins involved in mitochondrial DNA maintenance [transcription factor A (Tfam)], import (Tim23), and remodeling [mitofusin 2 (Mfn2) and dynamin-related protein 1 (Drp1)] did not parallel the decrease in mitochondrial content in PGC-1alpha KO animals. These proteins remained unchanged or were upregulated (P < 0.05) in the highly oxidative heart, indicating a change in mitochondrial composition. A change in muscle organelle composition was also evident from the alterations in subsarcolemmal and intermyofibrillar mitochondrial respiration, which was impaired in the absence of PGC-1alpha. However, endurance-trained KO animals did not exhibit reduced mitochondrial respiration. Mitochondrial reactive oxygen species (ROS) production was not affected by the lack of PGC-1alpha, but subsarcolemmal mitochondria from PGC-1alpha KO animals released a greater amount of cytochrome c than in WT animals following exogenous ROS treatment. Our results indicate that the lack of PGC-1alpha results in 1) a muscle type-specific suppression of mitochondrial content that depends on basal oxidative capacity, 2) an alteration in mitochondrial composition, 3) impaired mitochondrial respiratory function that can be improved by training, and 4) a greater basal protein release from subsarcolemmal mitochondria, indicating an enhanced mitochondrial apoptotic susceptibility.


Subject(s)
Apoptosis , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Physical Exertion , Trans-Activators/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Respiration , DNA, Mitochondrial/metabolism , Electron Transport Complex IV/metabolism , Hydrogen Peroxide/metabolism , Kinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Muscle/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proteins/metabolism , Muscle, Skeletal/pathology , Myocardium/pathology , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Endurance , Reactive Oxygen Species/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors
15.
Physiol Genomics ; 37(1): 58-66, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19106183

ABSTRACT

p53 is a tumor suppressor protein that also plays a role in regulating aerobic metabolism. Since skeletal muscle is a major source of whole body aerobic respiration, it is important to delineate the effects of p53 on muscle metabolism. In p53 knockout (KO) mice, we observed diminished mitochondrial content in mixed muscle and lowered peroxisome proliferator-activated receptor-gamma (PPARgamma) coactivator (PGC)-1alpha protein levels in gastrocnemius muscle. In intermyofibrillar (IMF) mitochondria, lack of p53 was associated with reduced respiration and elevated reactive oxygen species production. Permeability transition pore kinetics remained unchanged; however, IMF mitochondrial cytochrome c release was reduced and DNA fragmentation was lowered, illustrating a resistance to mitochondrially driven apoptosis in muscle of KO mice. p53-null animals displayed similar muscle strength but greater fatigability and less locomotory endurance than wild-type (WT) animals. Surprisingly, the adaptive responses in mitochondrial content to running were similar in WT and KO mice. Thus p53 may be important, but not necessary, for exercise-induced mitochondrial biogenesis. In WT animals, acute muscle contractions induced the phosphorylation of p53 in concert with increased activation of upstream kinases AMP-activated protein kinase and p38, indicating a pathway through which p53 may initiate mitochondrial biogenesis in response to contractile activity. These data illustrate a novel role for p53 in maintaining mitochondrial biogenesis, apoptosis, and performance in skeletal muscle.


Subject(s)
Apoptosis , Mitochondria/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Biomarkers/metabolism , Body Weight , Fatigue/physiopathology , Kinetics , Mice , Mice, Knockout , Mitochondria/pathology , Mitochondria/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Muscle Contraction , Muscle, Skeletal/physiopathology , Muscle, Skeletal/ultrastructure , Organ Size , Phosphoserine/metabolism , Physical Conditioning, Animal
16.
Aging (Albany NY) ; 1(9): 818-30, 2009 Sep 12.
Article in English | MEDLINE | ID: mdl-20157569

ABSTRACT

Our intent was to investigate the mechanisms driving the adaptive potential of subsarcolemmal (SS) and intermyofibrillar (IMF) mitochondria in young (6 mo) and senescent (36 mo) animals in response to a potent stimulus for organelle biogenesis. We employed chronic electrical stimulation (10 Hz, 3 h/day, 7 days) to induce contractile activity of skeletal muscle in 6 and 36 mo F344XBN rats. Subsequent to chronic activity, acute stimulation (1 Hz, 5 min) in situ revealed greater fatigue resistance in both age groups. However, the improvement in endurance was significantly greater in the young, compared to the old animals. Chronic muscle use also augmented SS and IMF mitochondrial volume to a greater extent in young muscle. The molecular basis for the diminished organelle expansion in aged muscle was due, in part, to the collective attenuation of the chronic stimulation-evoked increase in regulatory proteins involved in mediating mitochondrial protein import and biogenesis. Furthermore, adaptations in mitochondrial function were also blunted in old animals. However, chronic contractile activity evoked greater reductions in mitochondrially-mediated proapoptotic signaling in aged muscle. Thus, mitochondrial plasticity is retained in aged animals, however the magnitude of the changes are less compared to young animals due to attenuated molecular processes regulating organelle biogenesis.


Subject(s)
Adaptation, Psychological/physiology , Aging/physiology , Mitochondria, Muscle/physiology , Muscle, Skeletal/physiology , Animals , Apoptosis/physiology , DNA Fragmentation , Electric Stimulation , Electron Transport Complex IV/metabolism , Male , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Size/physiology , Muscle Contraction/physiology , Muscle Fatigue/physiology , Muscle Proteins/metabolism , Muscle, Skeletal/anatomy & histology , Ornithine Carbamoyltransferase/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Protein Transport/physiology , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Succinic Acid/metabolism , Succinic Acid/pharmacology
17.
Neuromolecular Med ; 10(4): 275-90, 2008.
Article in English | MEDLINE | ID: mdl-19005780

ABSTRACT

Substantial evidence indicates bioenergetic dysfunction and mitochondrial impairment contribute either directly and/or indirectly to the pathogenesis of numerous neurodegenerative disorders. Treatment paradigms aimed at ameliorating this cellular energy deficit and/or improving mitochondrial function in these neurodegenerative disorders may prove to be useful as a therapeutic intervention. Creatine is a molecule that is produced both endogenously, and acquired exogenously through diet, and is an extremely important molecule that participates in buffering intracellular energy stores. Once creatine is transported into cells, creatine kinase catalyzes the reversible transphosphorylation of creatine via ATP to enhance the phosphocreatine energy pool. Creatine kinase enzymes are located at strategic intracellular sites to couple areas of high energy expenditure to the efficient regeneration of ATP. Thus, the creatine kinase/phosphocreatine system plays an integral role in energy buffering and overall cellular bioenergetics. Originally, exogenous creatine supplementation was widely used only as an ergogenic aid to increase the phosphocreatine pool within muscle to bolster athletic performance. However, the potential therapeutic value of creatine supplementation has recently been investigated with respect to various neurodegenerative disorders that have been associated with bioenergetic deficits as playing a role in disease etiology and/or progression which include; Alzheimer's, Parkinson's, amyotrophic lateral sclerosis (ALS), and Huntington's disease. This review discusses the contribution of mitochondria and bioenergetics to the progression of these neurodegenerative diseases and investigates the potential neuroprotective value of creatine supplementation in each of these neurological diseases. In summary, current literature suggests that exogenous creatine supplementation is most efficacious as a treatment paradigm in Huntington's and Parkinson's disease but appears to be less effective for ALS and Alzheimer's disease.


Subject(s)
Creatine/pharmacology , Energy Metabolism/drug effects , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Adenosine Triphosphate/metabolism , Animals , Creatine/metabolism , Creatine/therapeutic use , Dietary Supplements , Energy Metabolism/physiology , Humans , Mitochondria/metabolism , Mitochondrial Diseases/physiopathology , Neurodegenerative Diseases/physiopathology , Phosphocreatine/metabolism
18.
Exerc Sport Sci Rev ; 36(3): 116-21, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18580291

ABSTRACT

Apoptosis is an essential process that plays a critical role in both tissue development and maintenance. Apoptosis has been shown to be involved in skeletal muscle atrophy resulting from chronic muscular disuse, sarcopenia, and mitochondrial myopathies. Exercise may attenuate some of the proapoptotic adaptations that occur during these conditions. This review will focus on the factors influencing mitochondrially mediated apoptosis in skeletal muscle.


Subject(s)
Apoptosis/physiology , Mitochondria, Muscle/physiology , Muscle, Skeletal/metabolism , Humans , Muscular Atrophy/pathology
19.
Am J Physiol Endocrinol Metab ; 293(3): E672-80, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17551003

ABSTRACT

Mitochondrial myopathy patients (MMPs) have impaired oxidative phosphorylation and exercise intolerance. Endurance training of MMPs improves exercise tolerance, but also increases mutational load. To assess the regulation of mitochondrial content in MMPs, we measured proteins involved in 1) biogenesis, 2) oxidative stress, and 3) apoptosis in MMPs and healthy controls (HCs) both before and after endurance training. Before training, MMPs had a greater mitochondrial content, along with a 1.4-fold (P < 0.05) higher expression of the biogenesis regulator peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha). The DNA repair enzyme 8-oxoguanine DNA glycolase-1 (OGG-1), the antioxidant manganese superoxide dismutase (MnSOD), and the apoptotic proteins AIF and Bcl-2 were higher in MMPs compared with HCs. Aconitase, an enzyme sensitive to oxidative stress, was 52% lower (P < 0.05) in MMPs when calculated based on an estimate of mitochondrial volume and oxidative stress-induced protein modifications tended to be higher in MMPs compared with HCs. Endurance training (ET) induced increases in mitochondrial content in both HC subjects and MMPs, but there was no effect of training on the regulatory proteins Tfam or PGC-1alpha. In MMPs, training induced a selective reduction of OGG-1, an increase in MnSOD, and a reduction in aconitase activity. Thus, before training, MMPs exhibited an adaptive response of nuclear proteins indicative of a compensatory increase in mitochondrial content. Following training, several parallel adaptations occurred in MMPs and HCs, which may contribute to previously observed functional improvements of exercise in MMPs. However, our results indicate that muscle from MMPs may be exposed to greater levels of oxidative stress during the course of training. Further investigation is required to evaluate the long-term benefits of endurance training as a therapeutic intervention for mitochondrial myopathy patients.


Subject(s)
Apoproteins/metabolism , DNA, Mitochondrial/genetics , Exercise , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/physiopathology , Adult , Cell Differentiation , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Mitochondrial Diseases/pathology
20.
Am J Physiol Endocrinol Metab ; 292(3): E748-55, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17106065

ABSTRACT

Chronic contractile activity of skeletal muscle induces an increase in mitochondria located in proximity to the sarcolemma [subsarcolemmal (SS)] and in mitochondria interspersed between the myofibrils [intermyofibrillar (IMF)]. These are energetically favorable metabolic adaptations, but because mitochondria are also involved in apoptosis, we investigated the effect of chronic contractile activity on mitochondrially mediated apoptotic signaling in muscle. We hypothesized that chronic contractile activity would provide protection against mitochondrially mediated apoptosis despite an elevation in the expression of proapoptotic proteins. To induce mitochondrial biogenesis, we chronically stimulated (10 Hz; 3 h/day) rat muscle for 7 days. Chronic contractile activity did not alter the Bax/Bcl-2 ratio, an index of apoptotic susceptibility, and did not affect manganese superoxide dismutase levels. However, contractile activity increased antiapoptotic 70-kDa heat shock protein and apoptosis repressor with a caspase recruitment domain by 1.3- and 1.4-fold (P<0.05), respectively. Contractile activity elevated SS mitochondrial reactive oxygen species (ROS) production 1.4- and 1.9-fold (P<0.05) during states IV and III respiration, respectively, whereas IMF mitochondrial state IV ROS production was suppressed by 28% (P<0.05) and was unaffected during state III respiration. Following stimulation, exogenous ROS treatment produced less cytochrome c release (25-40%) from SS and IMF mitochondria, and also reduced apoptosis-inducing factor release (approximately 30%) from IMF mitochondria, despite higher inherent cytochrome c and apoptosis-inducing factor expression. Chronic contractile activity did not alter mitochondrial permeability transition pore (mtPTP) components in either subfraction. However, SS mitochondria exhibited a significant increase in the time to Vmax of mtPTP opening. Thus, chronic contractile activity induces predominantly antiapoptotic adaptations in both mitochondrial subfractions. Our data suggest the possibility that chronic contractile activity can exert a protective effect on mitochondrially mediated apoptosis in muscle.


Subject(s)
Apoptosis , Mitochondria, Muscle/physiology , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Respiration , Electric Stimulation , Male , Mitochondria, Muscle/metabolism , Models, Biological , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sarcolemma/metabolism , Sarcolemma/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...