Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biol Res Nurs ; 23(3): 541-549, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33251850

RESUMO

BACKGROUND: Leptin and adiponectin, two adipokines involved in glucose and lipid metabolism, have been linked to regulation of growth in early infancy, energy balance, and metabolic disorders in childhood. The aim of this study was to determine if concentrations of leptin and adiponectin could be measured reliably in infants' saliva, to evaluate the degree of agreement with infant serum levels, and to explore their association with infant feeding status. METHODS: A total of 34 infants were recruited after birth and followed for 20 weeks. After log-transformation of the values, a Bland-Altman graphical approach was used to summarize the direction of the difference between the serum and saliva values. Repeated measures mixed modeling was used to evaluate differences over time in these outcomes by feeding status. RESULTS: Mean concentration of salivary leptin and adiponectin in infants was 3.7 (SD = .8) ng/mL and 2.9 (SD = 0.7) ng/mL, respectively. The degree of agreement between serum and saliva for log-transformed leptin and adiponectin values were relatively robust, albeit with a non-zero bias between the two methods, given that serum values were greater than corresponding saliva values for both adipokines in all infants. Each of the four repeated measures mixed models (one for each adipokine measure) had a significant main effect; however, the interaction between time and feeding status was not significant in any of the models. CONCLUSION: This study demonstrated that leptin and adiponectin can be measured in infant saliva, but in some cases leptin concentrations may be more difficult to detect.


Assuntos
Adiponectina , Comportamento Alimentar , Fenômenos Fisiológicos da Nutrição do Lactente , Leptina , Adipocinas/metabolismo , Adiponectina/metabolismo , Metabolismo Energético , Humanos , Lactente , Leptina/metabolismo , Saliva/metabolismo
2.
Front Physiol ; 7: 686, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28119629

RESUMO

Patients with heart failure (HF) have diaphragm abnormalities that contribute to disease morbidity and mortality. Studies in animals suggest that reactive oxygen species (ROS) cause diaphragm abnormalities in HF. However, the effects of HF on ROS sources, antioxidant enzymes, and protein oxidation in the diaphragm of humans is unknown. NAD(P)H oxidase, especially the Nox2 isoform, is an important source of ROS in the diaphragm. Our main hypothesis was that diaphragm from patients with HF have heightened Nox2 expression and p47phox phosphorylation (marker of enzyme activation) that is associated with elevated protein oxidation. We collected diaphragm biopsies from patients with HF and brain-dead organ donors (controls). Diaphragm mRNA levels of Nox2 subunits were increased 2.5-4.6-fold over controls (p < 0.05). Patients also had increased protein levels of Nox2 subunits (p47phox, p22phox, and p67phox) and total p47phox phosphorylation, while phospho-to-total p47phox levels were unchanged. The antioxidant enzyme catalase was increased in patients, whereas glutathione peroxidase and superoxide dismutases were unchanged. Among markers of protein oxidation, carbonyls were increased by ~40% (p < 0.05) and 4-hydroxynonenal and 3-nitrotyrosines were unchanged in patients with HF. Overall, our findings suggest that Nox2 is an important source of ROS in the diaphragm of patients with HF and increases in levels of antioxidant enzymes are not sufficient to maintain normal redox homeostasis. The net outcome is elevated diaphragm protein oxidation that has been shown to cause weakness in animals.

3.
Front Physiol ; 6: 136, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25972815

RESUMO

AIMS: At the time of diagnosis, 60% of lung cancer patients present with cachexia, a severe wasting syndrome that increases morbidity and mortality. Tumors secrete multiple factors that contribute to cachectic muscle wasting, and not all of these factors have been identified. We used Orbitrap electrospray ionization mass spectrometry to identify novel cachexia-inducing candidates in media conditioned with Lewis lung carcinoma cells (LCM). RESULTS: One-hundred and 58 proteins were confirmed in three biological replicates. Thirty-three were identified as secreted proteins, including 14-3-3 proteins, which are highly conserved adaptor proteins known to have over 200 binding partners. We confirmed the presence of extracellular 14-3-3 proteins in LCM via western blot and discovered that LCM contained less 14-3-3 content than media conditioned with C2C12 myotubes. Using a neutralizing antibody, we depleted extracellular 14-3-3 proteins in myotube culture medium, which resulted in diminished myosin content. We identified the proposed receptor for 14-3-3 proteins, CD13, in differentiated C2C12 myotubes and found that inhibiting CD13 via Bestatin also resulted in diminished myosin content. CONCLUSIONS: Our novel findings show that extracellular 14-3-3 proteins may act as previously unidentified myokines and may signal via CD13 to help maintain muscle mass.

5.
Redox Biol ; 2: 910-20, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25180167

RESUMO

AIMS: Sphingolipid and oxidant signaling affect glucose uptake, atrophy, and force production of skeletal muscle similarly and both are stimulated by tumor necrosis factor (TNF), suggesting a connection between systems. Sphingolipid signaling is initiated by neutral sphingomyelinase (nSMase), a family of agonist-activated effector enzymes. Northern blot analyses suggest that nSMase3 may be a striated muscle-specific nSMase. The present study tested the hypothesis that nSMase3 protein is expressed in skeletal muscle and functions to regulate TNF-stimulated oxidant production. RESULTS: We demonstrate constitutive nSMase activity in skeletal muscles of healthy mice and humans and in differentiated C2C12 myotubes. nSMase3 (Smpd4 gene) mRNA is highly expressed in muscle. An nSMase3 protein doublet (88 and 85 kD) is derived from alternative mRNA splicing of exon 11. The proteins partition differently. The full-length 88 kD isoform (nSMase3a) fractionates with membrane proteins that are resistant to detergent extraction; the 85 kD isoform lacking exon 11 (nSMase3b) is more readily extracted and fractionates with detergent soluble membrane proteins; neither variant is detected in the cytosol. By immunofluorescence microscopy, nSMase3 resides in both internal and sarcolemmal membranes. Finally, myotube nSMase activity and cytosolic oxidant activity are stimulated by TNF. Both if these responses are inhibited by nSMase3 knockdown. INNOVATION: These findings identify nSMase3 as an intermediate that links TNF receptor activation, sphingolipid signaling, and skeletal muscle oxidant production. CONCLUSION: Our data show that nSMase3 acts as a signaling nSMase in skeletal muscle that is essential for TNF-stimulated oxidant activity.


Assuntos
Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Oxidantes/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Linhagem Celular , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução
6.
J Appl Physiol (1985) ; 117(8): 921-9, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25170071

RESUMO

Recent work indicates that infections are a major contributor to diaphragm weakness in patients who are critically ill and mechanically ventilated, and that diaphragm weakness is a risk factor for death and prolonged mechanical ventilation. Infections activate muscle calpain, but many believe this is an epiphenomenon and that other proteolytic processes are responsible for infection-induced muscle weakness. We tested the hypothesis that muscle-specific overexpression of calpastatin (CalpOX; an endogenous calpain inhibitor) would attenuate diaphragm dysfunction in cecal ligation puncture (CLP)-induced sepsis. We studied 1) wild-type (WT) sham-operated mice, 2) WT CLP-operated mice, 3) CalpOX sham-operated mice, and 4) CalpOX CLP-operated mice (n = 9-10/group). Twenty-four hours after surgery, we assessed the diaphragm force-frequency relationship, diaphragm mass, and total protein content and diaphragm levels of talin and myosin heavy chain (MHC). CLP markedly reduced diaphragm-specific force generation (force/cross-sectional area), which was prevented by calpastatin overexpression (force averaged 21.4 ± 0.5, 6.9 ± 0.8, 22.4 ± 1.0, and 18.3 ± 1.3 N/cm(2), respectively, for WT sham, WT CLP, CalpOX sham, and CalpOX CLP groups, P < 0.001). Diaphragm mass and total protein content were similar in all groups. CLP induced talin cleavage and reduced MHC levels; CalpOX prevented these alterations. CLP-induced sepsis rapidly reduces diaphragm-specific force generation and is associated with cleavage and/or depletion of key muscle proteins (talin, MHC), effects prevented by muscle-specific calpastatin overexpression. These data indicate that calpain activation is a major cause of diaphragm weakness in response to CLP-induced sepsis.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Ceco/metabolismo , Diafragma/metabolismo , Diafragma/patologia , Debilidade Muscular/metabolismo , Músculos/metabolismo , Sepse/metabolismo , Animais , Calpaína/metabolismo , Ceco/patologia , Ligadura/métodos , Camundongos , Proteínas Musculares/metabolismo , Debilidade Muscular/patologia , Músculos/patologia , Cadeias Pesadas de Miosina/metabolismo , Sepse/patologia , Talina/metabolismo
7.
Front Physiol ; 5: 503, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25566096

RESUMO

AIMS: Cancer cachexia is a syndrome which results in severe loss of muscle mass and marked fatigue. Conditioned media from cachexia-inducing cancer cells triggers metabolic dysfunction in skeletal muscle, including decreased mitochondrial respiration, which may contribute to fatigue. We hypothesized that Lewis lung carcinoma conditioned medium (LCM) would impair the mitochondrial electron transport chain (ETC) and increase production of reactive oxygen species, ultimately leading to decreased mitochondrial respiration. We incubated C2C12 myotubes with LCM for 30 min, 2, 4, 24 or 48 h. We measured protein content by western blot; oxidant production by 2',7'-dichlorofluorescin diacetate (DCF), 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF), and MitoSox; cytochrome c oxidase activity by oxidation of cytochrome c substrate; and oxygen consumption rate (OCR) of intact myotubes by Seahorse XF Analyzer. RESULTS: LCM treatment for 2 or 24 h decreased basal OCR and ATP-related OCR, but did not alter the content of mitochondrial complexes I, III, IV and V. LCM treatment caused a transient rise in reactive oxygen species (ROS). In particular, mitochondrial superoxide (MitoSOX) was elevated at 2 h. 4-Hydroxynonenal, a marker of oxidative stress, was elevated in both cytosolic and mitochondrial fractions of cell lysates after LCM treatment. CONCLUSION: These data show that lung cancer-conditioned media alters electron flow in the ETC and increases mitochondrial ROS production, both of which may ultimately impair aerobic metabolism and decrease muscle endurance.

8.
J Appl Physiol (1985) ; 114(11): 1629-36, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23558387

RESUMO

TNF promotes skeletal muscle weakness, in part, by depressing specific force of muscle fibers. This is a rapid, receptor-mediated response, in which TNF stimulates cellular oxidant production, causing myofilament dysfunction. The oxidants appear to include nitric oxide (NO); otherwise, the redox mechanisms that underlie this response remain undefined. The current study tested the hypotheses that 1) TNF signals via neuronal-type NO synthase (nNOS) to depress specific force, and 2) muscle-derived reactive oxygen species (ROS) are essential co-mediators of this response. Mouse diaphragm fiber bundles were studied using live cell assays. TNF exposure increased general oxidant activity (P < 0.05; 2',7'-dichlorodihydrofluorescein diacetate assay) and NO activity (P < 0.05; 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate assay) and depressed specific force across the full range of stimulus frequencies (1-300 Hz; P < 0.05). These responses were abolished by pretreatment with N(ω)-nitro-L-arginine methyl ester (L-NAME; a nonspecific inhibitor of NOS activity), confirming NO involvement. Genetic nNOS deficiency replicated L-NAME effects on TNF-treated muscle, diminishing NO activity (-80%; P < 0.05) and preventing the decrement in specific force (P < 0.05). Comparable protection was achieved by selective depletion of muscle-derived ROS. Pretreatment with either SOD (degrades superoxide anion) or catalase (degrades hydrogen peroxide) depressed oxidant activity in TNF-treated muscle and abolished the decrement in specific force. These findings indicate that TNF signals via nNOS to depress contractile function, a response that requires ROS and NO as obligate co-mediators.


Assuntos
Diafragma/fisiologia , Força Muscular/fisiologia , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Cultivadas , Diafragma/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia
9.
J Appl Physiol (1985) ; 112(9): 1538-45, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22362402

RESUMO

Diseases that result in muscle weakness, e.g., heart failure, are characterized by elevated sphingomyelinase (SMase) activity. In intact muscle, SMase increases oxidants that contribute to diminished muscle force. However, the source of oxidants, specific processes of muscle contraction that are dysfunctional, and biochemical changes underlying the weakness elicited by SMase remain unknown. We tested three hypotheses: 1) SMase-induced depression of muscle force is mediated by mitochondrial reactive oxygen species (ROS), 2) SMase depresses force and calcium sensitivity of the contractile apparatus, and 3) SMase promotes oxidation and phosphorylation of myofibrillar proteins. Our experiments included intact muscle bundles, permeabilized single fibers, and isolated myofibrillar proteins. The mitochondrial-targeted antioxidant d-Arg-2',6'-dimethyl-Tyr-Lys-Phe-NH(2), decreased cytosolic oxidants and protected intact muscle bundles from weakness stimulated by SMase. SMase depressed maximal calcium-activated force by 20% in permeabilized single fibers (in kN/m(2): control 117 ± 6; SMase 93 ± 8; P < 0.05). Calcium sensitivity of permeabilized single fibers decreased from 5.98 ± 0.03 (control) to 5.91 ± 0.02 (SMase; P < 0.05). Myofibrillar protein nitrotyrosines, carbonyls, and phosphorylation were unaltered by SMase. Our study shows that the fall in specific force of intact muscle elicited by SMase is mediated by mitochondrial ROS and can be attributed largely to dysfunction of the contractile apparatus.


Assuntos
Sinalização do Cálcio , Diafragma/enzimologia , Contração Muscular , Fibras Musculares Esqueléticas/enzimologia , Força Muscular , Esfingomielina Fosfodiesterase/metabolismo , Animais , Antioxidantes/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Permeabilidade da Membrana Celular , Citosol/enzimologia , Diafragma/citologia , Diafragma/efeitos dos fármacos , Estimulação Elétrica , Acoplamento Excitação-Contração , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Musculares/enzimologia , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Força Muscular/efeitos dos fármacos , Miofibrilas/enzimologia , Oxirredução , Estresse Oxidativo , Fosforilação , Carbonilação Proteica , Processamento de Proteína Pós-Traducional , Espécies Reativas de Oxigênio/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
10.
Am J Physiol Cell Physiol ; 302(1): C195-202, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21940668

RESUMO

Doxorubicin, a commonly prescribed chemotherapeutic agent, causes skeletal muscle wasting in cancer patients undergoing treatment and increases mitochondrial reactive oxygen species (ROS) production. ROS stimulate protein degradation in muscle by activating proteolytic systems that include caspase-3 and the ubiquitin-proteasome pathway. We hypothesized that doxorubicin causes skeletal muscle catabolism through ROS, causing upregulation of E3 ubiquitin ligases and caspase-3. We tested this hypothesis by exposing differentiated C2C12 myotubes to doxorubicin (0.2 µM). Doxorubicin decreased myotube width 48 h following exposure, along with a 40-50% reduction in myosin and sarcomeric actin. Cytosolic oxidant activity was elevated in myotubes 2 h following doxorubicin exposure. This increase in oxidants was followed by an increase in the E3 ubiquitin ligase atrogin-1/muscle atrophy F-box (MAFbx) and caspase-3. Treating myotubes with SS31 (opposes mitochondrial ROS) inhibited expression of ROS-sensitive atrogin-1/MAFbx and protected against doxorubicin-stimulated catabolism. These findings suggest doxorubicin acts via mitochondrial ROS to stimulate myotube atrophy.


Assuntos
Doxorrubicina/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Humanos , Metabolismo/efeitos dos fármacos , Metabolismo/fisiologia , Fibras Musculares Esqueléticas/citologia
11.
Antioxid Redox Signal ; 15(9): 2465-75, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21453198

RESUMO

UNLABELLED: Skeletal muscle expresses prion protein (PrP) that buffers oxidant activity in neurons. AIMS: We hypothesize that PrP deficiency would increase oxidant activity in skeletal muscle and alter redox-sensitive functions, including contraction and glucose uptake. We used real-time polymerase chain reaction and Western blot analysis to measure PrP mRNA and protein in human diaphragm, five murine muscles, and muscle-derived C2C12 cells. Effects of PrP deficiency were tested by comparing PrP-deficient mice versus wild-type mice and morpholino-knockdown versus vehicle-treated myotubes. Oxidant activity (dichlorofluorescin oxidation) and specific force were measured in murine diaphragm fiber bundles. RESULTS: PrP content differs among mouse muscles (gastrocnemius>extensor digitorum longus, EDL>tibialis anterior, TA; soleus>diaphragm) as does glycosylation (di-, mono-, nonglycosylated; gastrocnemius, EDL, TA=60%, 30%, 10%; soleus, 30%, 40%, 30%; diaphragm, 30%, 30%, 40%). PrP is predominantly di-glycosylated in human diaphragm. PrP deficiency decreases body weight (15%) and EDL mass (9%); increases cytosolic oxidant activity (fiber bundles, 36%; C2C12 myotubes, 7%); and depresses specific force (12%) in adult (8-12 mos) but not adolescent (2 mos) mice. INNOVATION: This study is the first to directly assess a role of prion protein in skeletal muscle function. CONCLUSIONS: PrP content varies among murine skeletal muscles and is essential for maintaining normal redox homeostasis, muscle size, and contractile function in adult animals.


Assuntos
Músculo Esquelético/metabolismo , Príons/metabolismo , Animais , Western Blotting , Linhagem Celular , Diafragma/metabolismo , Humanos , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Oxirredução , Príons/genética
12.
J Physiol ; 589(Pt 9): 2171-9, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21320886

RESUMO

Chronic inflammatory diseases such as heart failure, cancer and arthritis have secondary effects on skeletal muscle that cause weakness and exercise intolerance. These symptoms exacerbate illness and make death more likely. Weakness is not simply a matter of muscle atrophy. Functional studies show that contractile dysfunction, i.e. a reduction in specific force, makes an equally important contribution to overall weakness. The most clearly defined mediator of contractile dysfunction is tumour necrosis factor (TNF). TNF serum levels are elevated in chronic disease, correlate with muscle weakness, and are a predictor of morbidity and mortality. Research is beginning to unravel the mechanism by which TNF depresses specific force. TNF acts via the TNFR1 receptor subtype to depress force by increasing cytosolic oxidant activity. Oxidants depress myofibrillar function, decreasing specific force without altering calcium regulation or other aspects of myofibrillar mechanics. Beyond these concepts, the intracellular mechanisms that depress specific force remain undefined. We do not know the pathway by which receptor-ligand interaction stimulates oxidant production. Nor do we know the type(s) of oxidants stimulated by TNF, their intracellular source(s), or their molecular targets. Investigators in the field are pursuing these issues with the long-term goal of preserving muscle function in individuals afflicted by chronic disease.


Assuntos
Inflamação/metabolismo , Contração Muscular , Força Muscular , Debilidade Muscular/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Animais , Doença Crônica , Humanos , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Debilidade Muscular/patologia , Debilidade Muscular/fisiopatologia , Atrofia Muscular/patologia , Atrofia Muscular/fisiopatologia , Oxirredução , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
13.
Am J Physiol Lung Cell Mol Physiol ; 300(2): L225-31, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21097524

RESUMO

Doxorubicin, a common chemotherapeutic agent, causes respiratory muscle weakness in both patients and rodents. Tumor necrosis factor-α (TNF), a proinflammatory cytokine that depresses diaphragm force, is elevated following doxorubicin chemotherapy. TNF-induced diaphragm weakness is mediated through TNF type 1 receptor (TNFR1). These findings lead us to hypothesize that TNF/TNFR1 signaling mediates doxorubicin-induced diaphragm muscle weakness. We tested this hypothesis by treating C57BL/6 mice with a clinical dose of doxorubicin (20 mg/kg) via intravenous injection. Three days later, we measured contractile properties of muscle fiber bundles isolated from the diaphragm. We tested the involvement of TNF/TNFR1 signaling using pharmaceutical and genetic interventions. Etanercept, a soluble TNF receptor, and TNFR1 deficiency protected against the depression in diaphragm-specific force caused by doxorubicin. Doxorubicin stimulated an increase in TNFR1 mRNA and protein (P < 0.05) in the diaphragm, along with colocalization of TNFR1 to the plasma membrane. These results suggest that doxorubicin increases diaphragm sensitivity to TNF by upregulating TNFR1, thereby causing respiratory muscle weakness.


Assuntos
Diafragma/efeitos dos fármacos , Diafragma/fisiopatologia , Doxorrubicina/efeitos adversos , Debilidade Muscular/induzido quimicamente , Debilidade Muscular/fisiopatologia , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Antineoplásicos/efeitos adversos , Sequência de Bases , Primers do DNA/genética , Etanercepte , Imunoglobulina G/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Debilidade Muscular/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética , Regulação para Cima/efeitos dos fármacos
14.
Muscle Nerve ; 43(1): 94-102, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21171100

RESUMO

Doxorubicin is a chemotherapeutic agent prescribed for a variety of tumors. While undergoing treatment, patients exhibit frequent symptoms that suggest respiratory muscle weakness. Cancer patients can receive doxorubicin chemotherapy through either intravenous (IV) or intraperitoneal (IP) injections. We hypothesized that respiratory muscle function would be depressed in a murine model of chemotherapy. We tested this hypothesis by treating C57BL/6 mice with a clinical dose of doxorubicin (20 mg/kg) via IV or IP injection. Three days later we measured contractile properties of muscle fiber bundles isolated from the diaphragm. Doxorubicin consistently depressed diaphragm force with both methods of administration (P < 0.01). Doxorubicin IP exaggerated the depression in diaphragm force and stimulated tissue inflammation and muscle fiber injury. These results suggest that clinically relevant doses of doxorubicin cause respiratory muscle weakness and that the loss of function depends, in part, on the route of administration.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Diafragma/efeitos dos fármacos , Modelos Animais de Doenças , Doxorrubicina/toxicidade , Debilidade Muscular/induzido quimicamente , Paralisia Respiratória/induzido quimicamente , Animais , Diafragma/patologia , Diafragma/fisiopatologia , Injeções Intraperitoneais/efeitos adversos , Injeções Intravenosas/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Debilidade Muscular/patologia , Debilidade Muscular/fisiopatologia , Paralisia Respiratória/patologia , Paralisia Respiratória/fisiopatologia
15.
Am J Physiol Cell Physiol ; 299(3): C552-60, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20519448

RESUMO

Sphingomyelinase (SMase) hydrolyzes membrane sphingomyelin into ceramide, which increases oxidants in nonmuscle cells. Serum SMase activity is elevated in sepsis and heart failure, conditions where muscle oxidants are increased, maximal muscle force is diminished, and fatigue is accelerated. We tested the hypotheses that exogenous SMase and accumulation of ceramide in muscle increases oxidants in muscle cells, depresses specific force of unfatigued muscle, and accelerates the fatigue process. We also anticipated that the antioxidant N-acetylcysteine (NAC) would prevent SMase effects on muscle function. We studied the responses of C2C12 myotubes and mouse diaphragm to SMase treatment in vitro. We observed that SMase caused a 2.8-fold increase in total ceramide levels in myotubes. Exogenous ceramide and SMase elevated oxidant activity in C2C12 myotubes by 15-35% (P < 0.05) and in diaphragm muscle fiber bundles by 58-120% (P < 0.05). The SMase-induced increase in diaphragm oxidant activity was prevented by NAC. Exogenous ceramide depressed diaphragm force by 55% (P < 0.05), while SMase depressed maximal force by 30% (P < 0.05) and accelerated fatigue--effects opposed by treatment with NAC. In conclusion, our findings suggest that SMase stimulates a ceramide-oxidant signaling pathway that results in muscle weakness and fatigue.


Assuntos
Fadiga Muscular/fisiologia , Músculo Esquelético/fisiologia , Oxidantes/fisiologia , Esfingomielina Fosfodiesterase/fisiologia , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Proteínas de Bactérias/farmacologia , Linhagem Celular , Ceramidas/metabolismo , Citosol/metabolismo , Diafragma/fisiologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular , Fibras Musculares Esqueléticas/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Transdução de Sinais , Esfingomielina Fosfodiesterase/farmacologia
16.
J Appl Physiol (1985) ; 107(6): 1935-42, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19779154

RESUMO

Cancer patients receiving doxorubicin chemotherapy experience both muscle weakness and fatigue. One postulated mediator of the muscle dysfunction is an increase in tumor necrosis factor-alpha (TNF), a proinflammatory cytokine that mediates limb muscle contractile dysfunction through the TNF receptor subtype 1 (TNFR1). Our main hypothesis was that systemic doxorubicin administration would cause muscle weakness and fatigue. Systemic doxorubicin administration (20 mg/kg) depressed maximal force of the extensor digitorum longus (EDL; P < 0.01), accelerated EDL fatigue (P < 0.01), and elevated serum TNF levels (P < 0.05) 72 h postinjection. Genetic TNFR1 deficiency prevented the fall in specific force caused by systemic doxorubicin, without protecting against fatigue (P < 0.01). These results demonstrate that clinical doxorubicin concentrations disrupt limb muscle function in a TNFR1-dependent manner.


Assuntos
Doxorrubicina/administração & dosagem , Contração Muscular/efeitos dos fármacos , Força Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Análise de Variância , Animais , Western Blotting , Cálcio/metabolismo , Ensaio de Imunoadsorção Enzimática , Masculino , Camundongos , Camundongos Knockout , Fadiga Muscular/efeitos dos fármacos , Debilidade Muscular/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Músculo Esquelético/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
17.
Am J Physiol Cell Physiol ; 297(3): C706-14, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19625606

RESUMO

Interleukin-1 (IL-1) is an inflammatory cytokine that has been linked to muscle catabolism, a process regulated by muscle-specific E3 proteins of the ubiquitin-proteasome pathway. To address cellular mechanism, we tested the hypothesis that IL-1 induces myofibrillar protein loss by acting directly on muscle to increase expression of two critical E3 proteins, atrogin1/muscle atrophy F-box (MAFbx) and muscle RING-finger 1 (MuRF1). Experiments were conducted using mature C2C12 myotubes to eliminate systemic cytokine effects and avoid paracrine signaling by nonmuscle cell types. Time-course protocols were used to define the sequence of cellular responses. We found that atrogin1/MAFbx mRNA and MuRF1 mRNA are elevated 60-120 min after myotube exposure to either IL-1alpha or IL-1beta. These responses are preceded by signaling events that promote E3 expression. Both IL-1 isoforms stimulate phosphorylation of p38 mitogen-activated protein kinase and stimulate nuclear factor-kappaB (NF-kappaB) signaling; I-kappaB levels fall and NF-kappaB DNA binding activity increases. Other regulators of E3 expression are unaffected by IL-1 [cytosolic oxidant activity, Forkhead-O (Foxo) activity] or respond paradoxically (AKT). Chronic exposure of C2C12 myotubes over 48 h resulted in reduced myotube width and loss of sarcomeric actin. We conclude that IL-1alpha and IL-1beta act via an oxidant- and AKT/Foxo-independent mechanism to activate p38 MAPK, stimulate NF-kappaB signaling, increase expression of atrogin1/MAFbx and MuRF1, and reduce myofibrillar protein in differentiated myotubes.


Assuntos
Interleucina-1alfa/farmacologia , Interleucina-1beta/farmacologia , Fibras Musculares Esqueléticas/metabolismo , Animais , Linhagem Celular , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Proteínas Musculares/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
J Physiol ; 587(Pt 13): 3363-73, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19403598

RESUMO

Alternatives to the canonical insulin-stimulated pathway for glucose uptake are exercise- and exogenous reactive oxygen species (ROS)-stimulated glucose uptake. We proposed a model wherein mechanical loading, i.e. stretch, stimulates production of ROS to activate AMP-activated kinase (AMPK) to increase glucose uptake. Immunoblotting was used to measure protein phosphorylation; the fluorochrome probe 2'7'-dichlorofluorescin diacetate was used to measure cytosolic oxidant activity and 2-deoxy-d[1,2-(3)H]glucose was used to measure glucose uptake. The current studies demonstrate that stretch increases ROS, AMPKalpha phosphorylation and glucose transport in murine extensor digitorum longus (EDL) muscle (+121%, +164% and +184%, respectively; P < 0.05). We also demonstrate that stretch-induced glucose uptake persists in transgenic mice expressing an inactive form of the AMPKalpha2 catalytic subunit in skeletal muscle (+173%; P < 0.05). MnTBAP, a superoxide dismutase (SOD) mimetic, N-acteyl cysteine (NAC), a non-specific antioxidant, ebselen, a glutathione mimetic, or combined SOD plus catalase (ROS-selective scavengers) all decrease stretch-stimulated glucose uptake (P < 0.05) without changing basal uptake (P > 0.16). We also demonstrate that stretch-stimulated glucose uptake persists in the presence of the phosphatidylinositol 3-kinase (PI3-K) inhibitors wortmannin and LY294001 (P < 0.05) but is diminished by the p38-MAPK inhibitors SB203580 and A304000 (P > 0.99). These data indicate that stretch-stimulated glucose uptake in skeletal muscle is mediated by a ROS- and p38 MAPK-dependent mechanism that appears to be AMPKalpha2- and PI3-K-independent.


Assuntos
Glucose/metabolismo , Músculo Esquelético/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por AMP/deficiência , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Transporte Biológico Ativo , Células Cultivadas , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Contração Muscular/fisiologia , Músculo Esquelético/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Estresse Mecânico
19.
Crit Care Med ; 37(10 Suppl): S337-46, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20046119

RESUMO

Patients in the intensive care unit commonly develop muscle weakness. In part, this reflects loss of mechanical loading due to physical inactivity, bed rest, or immobilization. Mechanical unloading stimulates a complex adaptive response that results in muscle atrophy and loss of specific force. One element of this response is slowing of protein synthesis, which is regulated by signaling pathways downstream of mammalian target of rapamycin and insulin-like growth factor-1. In parallel, protein degradation is accelerated via three coordinate processes: calcium-dependent proteolysis, adenosine triphosphate-dependent proteolysis, and lysosomal proteolysis. Finally, unloading stimulates apoptosis of a subset of myonuclei within multinucleated muscle fibers. This helps to stabilize the relationship between nuclear number and cell volume during atrophy. Each of these responses is promoted by concurrent development of oxidative stress caused by increased production of reactive oxygen species in unloaded muscle fibers. Countermeasures that lessen the effects of unloading include physical activity, nutritional supplements, hormone therapy, and antioxidant administration. Targeted research is needed to define the role of mechanical unloading in intensive care unit-associated weakness and develop countermeasures to preserve muscle function, lessen illness, and hasten the recovery of critically ill patients.


Assuntos
Repouso em Cama/efeitos adversos , Unidades de Terapia Intensiva , Debilidade Muscular/reabilitação , Músculo Esquelético/fisiopatologia , Antioxidantes/administração & dosagem , Cuidados Críticos/métodos , Estado Terminal/reabilitação , Suplementos Nutricionais , Exercício Físico , Humanos , Movimento , Debilidade Muscular/etiologia , Atrofia Muscular/etiologia , Atrofia Muscular/reabilitação
20.
Am J Physiol Cell Physiol ; 295(4): C986-93, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18701653

RESUMO

Murine models of starvation-induced muscle atrophy demonstrate that reduced protein kinase B (AKT) function upregulates the atrophy-related gene atrogin-1/MAFbx (atrogin). The mechanism involves release of inhibition of Forkhead transcription factors, namely Foxo1 and Foxo3. Elevated atrogin mRNA also corresponds with elevated TNF in inflammatory catabolic states, including cancer and chronic heart failure. Exogenous tumor necrosis factor (TNF) increases atrogin mRNA in vivo and in vitro. We used TNF-treated C2C12 myotubes to test the hypothesis that AKT-Foxo1/3 signaling mediates TNF regulation of atrogin mRNA. Here we confirm that exposure to TNF increases atrogin mRNA (+125%). We also confirm that canonical AKT-mediated regulation of atrogin is active in C2C12 myotubes. Inhibition of phosphoinositol-3 kinase (PI3K)/AKT signaling with wortmannin reduces AKT phosphorylation (-87%) and increases atrogin mRNA (+340%). Activation with insulin-like growth factor (IGF) increases AKT phosphorylation (+126%) and reduces atrogin mRNA (-15%). Although AKT regulation is intact, our data suggest it does not mediate TNF effects on atrogin. TNF increases AKT phosphorylation (+50%) and stimulation of AKT with IGF does not prevent TNF induction of atrogin mRNA. Nor does TNF appear to signal through Foxo1/3 proteins. TNF has no effect on Foxo1/3 mRNA or Foxo1/3 nuclear localization. Instead, TNF increases nuclear Foxo4 protein (+55%). Small interfering RNA oligos targeted to two distinct regions of Foxo4 mRNA reduce the TNF-induced increase in atrogin mRNA (-34% and -32%). We conclude that TNF increases atrogin mRNA independent of AKT via Foxo4. These results suggest a mechanism by which inflammatory catabolic states may persist in the presence of adequate growth factors and nutrition.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteínas Musculares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Camundongos , Proteínas Musculares/genética , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Isoformas de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Transdução de Sinais , Somatomedinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA