Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Exerc Sport Sci Rev ; 48(3): 140-148, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32568926

RESUMEN

We review evidence supporting an updated mechanostat model in bone that highlights the central role of osteocytes within bone's four mechanoadaptive pathways: 1) formation modeling and 2) targeted remodeling, which occur with heightened mechanical loading, 3) resorption modeling, and 4) disuse-mediated remodeling, which occur with disuse. These four pathways regulate whole-bone stiffness in response to changing mechanical demands.


Asunto(s)
Regeneración Ósea , Resorción Ósea , Osteocitos/fisiología , Adaptación Fisiológica , Animales , Fenómenos Biomecánicos , Hueso Cortical/fisiología , Humanos , Estrés Mecánico , Soporte de Peso/fisiología
2.
BMC Musculoskelet Disord ; 20(1): 282, 2019 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-31185965

RESUMEN

BACKGROUND: Musculoskeletal injuries (MSKIs) are common in military trainees and present a considerable threat to occupational fitness, deployability, and overall military readiness. Despite the negative effects of MSKIs on military readiness, comprehensive evaluations of the key known and possible risk factors for MSKIs are lacking. The U.S. Army Research Institute of Environmental Medicine (ARIEM) is initiating a large-scale research effort, the ARIEM Reduction in Musculoskeletal Injury (ARMI) Study, to better understand the interrelationships among a wide range of potential MSKI risk factors in U.S. Army trainees in order to identify those risk factors that most contribute to MSKI and may be best targeted for effective mitigation strategies. METHODS: This prospective study aims to enroll approximately 4000 (2000 male and 2000 female) U.S. Army trainees undergoing Basic Combat Training (BCT). Comprehensive in-person assessments will be completed at both the beginning and end of BCT. Participants will be asked to complete surveys of personal background information, medical history, physical activity, sleep behaviors, and personality traits. Physical measurements will be performed to assess anthropometrics, tibial microarchitecture and whole body bone mineral density, muscle cross-sectional area, body composition, and muscle function. Blood sampling will be also be conducted to assess musculoskeletal, genetic, and nutritional biomarkers of risk. In addition, participants will complete weekly surveys during BCT that examine MSKI events, lost training time, and discrete risk factors for injury. Participants' medical records will be tracked for the 2 years following graduation from training to identify MSKI events and related information. Research hypotheses focus on the development of a multivariate prediction model for MSKI. DISCUSSION: Results from this study are expected to inform current understanding of known and potential risk factors for MSKIs that can be incorporated into solutions that optimize Soldier health and enhance military readiness.


Asunto(s)
Ejercicio Físico/fisiología , Personal Militar/estadística & datos numéricos , Enfermedades Musculoesqueléticas/epidemiología , Sistema Musculoesquelético/lesiones , Adolescente , Adulto , Diseño de Investigaciones Epidemiológicas , Femenino , Humanos , Estudios Longitudinales , Masculino , Enfermedades Musculoesqueléticas/fisiopatología , Enfermedades Musculoesqueléticas/prevención & control , Sistema Musculoesquelético/fisiopatología , Estudios Prospectivos , Factores de Riesgo , Estados Unidos/epidemiología , Adulto Joven
3.
Biochem Biophys Res Commun ; 482(4): 1420-1426, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27965101

RESUMEN

Skeletal muscle metabolic homeostasis is maintained through numerous biochemical and physiological processes. Two principal molecular regulators of skeletal muscle metabolism include AMP-activated protein kinase (AMPK) and phosphatidylinositol 3-kinase (PI3K); however, PI3K exists as multiple isoforms, and specific metabolic actions of each isoform have not yet been fully elucidated in skeletal muscle. Given this lack of knowledge, we performed a series of experiments to define the extent to which PI3K p110ß mediated expression and (or) activation of AMPK in skeletal muscle. To determine the effect of p110ß inhibition on AMPK expression and phosphorylation in cultured cells, C2C12 myoblasts were treated with a pharmacological inhibitor of p110ß (TGX-221), siRNA against p110ß, or overexpression of kinase-dead p110ß. Expression and phosphorylation of AMPK were unaffected in myoblasts treated with TGX-221 or expressing kinase-dead p110ß. However, expressions of total and phosphorylated AMPK at T172 were reduced in myoblasts treated with p110ß siRNA. When normalized to expression of total AMPK, phosphorylation of AMPK S485/491 was elevated in p110ß-deficient myoblasts. Similar results were observed in tibialis anterior muscle from mice with conditional deletion of p110ß (p110ß-mKO mice). Analysis of AMPK transcript expression revealed decreased expression of Prkaa2 in p110ß-deficient myoblasts and in p110ß-mKO muscle. Loss of p110ß had no effect on oligomycin-stimulated phosphorylation of AMPK or phosphorylated Acetyl-CoA carboxylase (ACC), although oligomycin-induced AMPK and ACC phosphorylation were increased in p110ß-deficient myoblasts compared to oligomycin-stimulated control myoblasts when normalized to levels of total AMPK or ACC. Overall, these results suggest that p110ß positively regulates expression of AMPK in cultured myoblasts and in skeletal muscle in vivo; moreover, despite the reduced abundance of AMPK in p110ß-deficient myoblasts, loss of p110ß does not appear to impair AMPK activation following stimulus. These findings thus reveal a novel role for p110ß in mediating skeletal muscle metabolic signaling.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Regulación de la Expresión Génica , ARN Interferente Pequeño/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Catálisis , Línea Celular , Eliminación de Gen , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculos/metabolismo , Mioblastos/metabolismo , Fosforilación
4.
Biochem Biophys Res Commun ; 469(4): 1117-22, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26742424

RESUMEN

AMP-activated protein kinase (AMPK) is a serine/threonine kinase that functions as a sensor of intracellular energy. Activation of AMPK is associated with increased phosphorylation of the α-subunit at threonine 172 (T172) and decreased phosphorylation at serine 485 in AMPKα1 and serine 491 in AMPKα2 (S485/491). One potential mediator of AMPK phosphorylation is phosphatidylinositol 3-kinase (PI3K); however, the mechanism and the identities of the specific PI3K isoforms that regulate AMPK activation are not known. To determine whether PI3K p110α regulated AMPK activation in muscle cells, C2C12 myoblasts were subjected to pharmacological inhibition of p110α, siRNA directed against p110α, or overexpression of constitutively-active or dominant negative p110α. Chemical inhibition, siRNA, and expression of dominant-negative p110α were all associated with increased AMPK T172 phosphorylation, whereas expression of constitutively-active p110α reduced T172 phosphorylation. Conversely, pharmacological inhibition of p110α reduced AMPK S485/491 phosphorylation, while constitutively-active p110α increased AMPK S485/491 phosphorylation. This p110α-mediated increase in AMPK S485/491 phosphorylation was eliminated in the presence of the Akt inhibitor MK2206, suggesting that p110α-mediated phosphorylation of AMPKα at S485/491 is Akt-dependent. In response to oligomycin or serum-starvation, AMPK T172 phosphorylation was elevated in p110α-deficient myoblasts compared to control myoblasts. Overall, our findings identify PI3K p110α as a mediator of AMPK phosphorylation in myoblasts.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Mioblastos/enzimología , Estrés Oxidativo/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Línea Celular , Fosfatidilinositol 3-Quinasa Clase I , Ratones , Fosforilación
5.
Eur J Appl Physiol ; 114(11): 2251-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25027064

RESUMEN

PURPOSE: To determine the effects of US Army Ranger Training, an 8-week, physically demanding program (energy expenditure of 2,500-4,500 kcal/day) with energy restriction (deficit of 1,000-4,000 kcal/day) and sleep deprivation (<4 h sleep/night) on bone metabolism. METHODS: Blood was collected from 22 men (age 24 ± 4 years) before and after training. Follow-up measurements were made in a subset of 8 subjects between 2 and 6 weeks after training. Serum was analyzed for bone formation biomarkers [bone alkaline phosphatase (BAP) and osteocalcin (OCN)], bone resorption biomarkers [C-telopeptide cross-links of type I collagen (CTX) and tartrate-resistant acid phosphatase (TRAP5b)], calcium, parathyroid hormone (PTH), and vitamin D 25(OH)D increased significantly by 37.3 ± 45.2 % with training [corrected]. A repeated-measures ANOVA with time as the only factor was used to analyze data on the subset of 8 subjects who completed follow-up data collection. RESULTS: BAP and OCN significantly decreased by 22.8 ± 15.5% (pre 41.9 ± 10.1; post 31.7 ± 7.8 ng/ml) and 21.0 ± 23.3% (pre 15.0 ± 3.5; post 11.3 ± 2.1 ng/ml), respectively, with training, suggesting suppressed bone formation. OCN returned to baseline, while BAP remained suppressed 2-6 weeks post-training. TRAP5b significantly increased by 57.5 ± 51.6% (pre 3.0 ± 0.9; post 4.6 ± 1.4 ng/ml) from pre- to post-training, suggesting increased bone resorption, and returned to baseline 2-6 weeks post-training. PTH Increased significantly by 37.3 ± 45.2% with training. No changes in CTX, calcium, or PTH were detected. CONCLUSIONS: These data indicate that multi-stressor military training results in increased bone resorption and suppressed bone formation, with recovery of bone metabolism 2-6 weeks after completion of training.


Asunto(s)
Personal Militar , Osteogénesis , Entrenamiento de Fuerza/efectos adversos , Estrés Fisiológico , Fosfatasa Ácida/sangre , Adulto , Fosfatasa Alcalina/sangre , Resorción Ósea/etiología , Restricción Calórica/efectos adversos , Colágeno Tipo I/sangre , Humanos , Isoenzimas/sangre , Masculino , Osteocalcina/sangre , Hormona Paratiroidea/sangre , Péptidos/sangre , Privación de Sueño/complicaciones , Fosfatasa Ácida Tartratorresistente , Vitamina D/sangre
6.
J Int Soc Sports Nutr ; 21(1): 2302046, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38198469

RESUMEN

BACKGROUND: Non-steroidal anti-inflammatory drugs (NSAIDs) like ibuprofen, flurbiprofen, naproxen sodium, and indomethacin are commonly employed for their pain-relieving and inflammation-reducing qualities. NSAIDs work by blocking COX-1 and/or COX-2, enzymes which play roles in inflammation, fever, and pain. The main difference among NSAIDs lies in their affinity to these enzymes, which in turn, influences prostaglandin secretion, and skeletal muscle growth and regeneration. The current study investigated the effects of NSAIDs on human skeletal muscle cells, focusing on myoblast proliferation, differentiation, and muscle protein synthesis signaling. METHODS: Using human primary muscle cells, we examined the dose-response impact of flurbiprofen (25-200 µM), indomethacin (25-200 µM), ibuprofen (25-200 µM), and naproxen sodium (25-200 µM), on myoblast viability, myotube area, fusion, and prostaglandin production. RESULTS: We found that supraphysiological concentrations of indomethacin inhibited myoblast proliferation (-74 ± 2% with 200 µM; -53 ± 3% with 100 µM; both p < 0.05) compared to control cells and impaired protein synthesis signaling pathways in myotubes, but only attenuated myotube fusion at the highest concentrations (-18 ± 2% with 200 µM, p < 0.05) compared to control myotubes. On the other hand, ibuprofen had no such effects. Naproxen sodium only increased cell proliferation at low concentrations (+36 ± 2% with 25 µM, p < 0.05), and flurbiprofen exhibited divergent impacts depending on the concentration whereby low concentrations improved cell proliferation (+17 ± 1% with 25 µM, p < 0.05) but high concentrations inhibited cell proliferation (-32 ± 1% with 200 µM, p < 0.05). CONCLUSION: Our findings suggest that indomethacin, at high concentrations, may detrimentally affect myoblast proliferation and differentiation via an AKT-dependent mechanism, and thus provide new understanding of NSAIDs' effects on skeletal muscle cell development.


Asunto(s)
Flurbiprofeno , Naproxeno , Humanos , Naproxeno/farmacología , Ibuprofeno/farmacología , Flurbiprofeno/farmacología , Indometacina/farmacología , Antiinflamatorios no Esteroideos/farmacología , Fibras Musculares Esqueléticas , Inflamación , Dolor , Prostaglandinas
7.
Med Sci Sports Exerc ; 56(9): 1644-1654, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38758530

RESUMEN

PURPOSE: The purpose of this study is to optimize a dMS-based urinary proteomic technique and evaluate the relationship between urinary proteome content and adaptive changes in bone microarchitecture during BCT. METHODS: Urinary proteomes were analyzed with an optimized dMS technique in two groups of 13 recruits ( N = 26) at the beginning (Pre) and end (Post) of BCT. Matched by age (21 ± 4 yr), sex (16 W), and baseline tibial trabecular bone volume fractions (Tb.BV/TV), these groups were distinguished by the most substantial (High) and minimal (Low) improvements in Tb.BV/TV. Differential protein expression was analyzed with mixed permutation ANOVA and false discovery proportion-based adjustment for multiple comparisons. RESULTS: Tibial Tb.BV/TV increased from pre- to post-BCT in High (3.30 ± 1.64%, P < 0.0001) but not Low (-0.35 ± 1.25%, P = 0.4707). The optimized dMS technique identified 10,431 peptides from 1368 protein groups that represented 165 integrative biological processes. Seventy-four urinary proteins changed from pre- to post-BCT ( P = 0.0019), and neutrophil-mediated immunity was the most prominent ontology. Two proteins (immunoglobulin heavy constant gamma 4 and C-type lectin domain family 4 member G) differed from pre- to post-BCT in High and Low ( P = 0.0006). CONCLUSIONS: The dMS technique can identify more than 1000 urinary proteins. At least 74 proteins are responsive to BCT, and other principally immune system-related proteins show differential expression patterns that coincide with adaptive bone formation.


Asunto(s)
Biomarcadores , Hueso Esponjoso , Personal Militar , Proteómica , Humanos , Masculino , Hueso Esponjoso/diagnóstico por imagen , Biomarcadores/orina , Adulto Joven , Tibia/metabolismo , Proteoma , Femenino , Adolescente
8.
J Recept Signal Transduct Res ; 33(1): 41-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23252604

RESUMEN

CONTEXT: G-protein coupled receptor (GPCR) signaling in skeletal muscle is incompletely understood; in particular, the signaling pathways that regulate GPCR-mediated signaling in skeletal muscle are only beginning to be established. Lysophosphatidic acid (LPA) is a GPCR agonist that has previously been shown to activate protein kinase D (PKD) in non-muscle cells; however, whether PKD is activated in response to LPA in skeletal muscle myoblasts, and the identities of signaling intermediates that regulate this activation, have not been defined. OBJECTIVE: To determine whether PKD is activated in response to LPA administration in myoblasts, and to define the signaling pathways that mediate LPA-stimulated PKD phosphorylation. METHODS: C2C12 myoblasts were treated with LPA and signaling pathways examined by means of Western immunoblotting and real-time PCR (RT-PCR). Pharmacological inhibition and RNA-interference were used to target specific molecules to determine their involvement in LPA-induced PKD phosphorylation. RESULTS: Treatment of myoblasts with exogenous LPA revealed that PI3K p110ß mediated PKD phosphorylation at Ser 748 and at Ser 916 through kinase-dependent and kinase-independent mechanisms. Loss of PKCδ, but not the loss of PKCα, prevented LPA-induced PKD phosphorylation. The PKD isoform responsive to LPA treatment was identified as PKD2. CONCLUSION: These results indicate that LPA-stimulated PKD2 phosphorylation requires PKCδ and non-catalytic actions of PI3K p110ß, and provide new information with respect to GPCR-mediated signal transduction in myoblasts.


Asunto(s)
Lisofosfolípidos/farmacología , Mioblastos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C-delta/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Línea Celular , Ratones , Mioblastos/efectos de los fármacos , Fosforilación/efectos de los fármacos
9.
Physiol Rep ; 11(20): e15805, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37879895

RESUMEN

AKT signaling plays a crucial role in muscle physiology, and is activated by stimuli, including insulin, growth factors, and exercise. Three AKT isoforms have been identified in mammals, and they possess both distinct and redundant functions. However, it is currently unknown what the predominant AKT isoform is in primary human skeletal myotubes, and very little is known regarding the effects of insulin and insulin-like growth factor-I (IGF-I) on AKT isoforms activation in human myotubes. Thus, we sought to determine the abundances of each AKT isoform in primary human skeletal myotubes and their responses to insulin or IGF-I. Analysis of protein lysates by liquid chromatography-parallel reaction monitoring/mass spectrometry revealed that AKT1 was the most abundant AKT isoform and AKT3 was the least-abundant isoform. Next, myotubes were treated with either 100 nM insulin or 10 nM IGF-I for 5, 20, 45, or 60 min. In response to insulin, there was a significant treatment effect on phosphorylation of AKT1 and AKT2, but not AKT3 (p < 0.01). In response to IGF-I, there was a significant treatment effect on phosphorylation of pan-AKT at all timepoints compared to control (p < 0.01). Next, we determined how much of the total AKT isoform pool was phosphorylated. For insulin stimulation, AKT1 was significantly higher than AKT2 at 5 min and 60 min posttreatment (p < 0.05 both) and significantly different than AKT3 at all timepoints (p < 0.05). For IGF-I stimulation, AKT1 was significantly higher than AKT2 at 45 and 60 min posttreatment (p < 0.05 both) and significantly higher than AKT3 at all timepoints (p < 0.05). Our findings reveal the differential phosphorylation patterns among the AKT isoforms and suggest a potential explanation for the regulatory role of AKT1 in skeletal muscle.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , Proteínas Proto-Oncogénicas c-akt , Animales , Humanos , Insulina/farmacología , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Mamíferos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fosforilación , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
10.
J Int Soc Sports Nutr ; 20(1): 2164209, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36620755

RESUMEN

Background: Cellular inflammatory response, mediated by arachidonic acid (AA) and cyclooxygenase, is a highly regulated process that leads to the repair of damaged tissue. Recent studies on murine C2C12 cells have demonstrated that AA supplementation leads to myotube hypertrophy. However, AA has not been tested on primary human muscle cells. Therefore, the purpose of this study was to determine whether AA supplementation has similar effects on human muscle cells. Methods: Proliferating and differentiating human myoblasts were exposed to AA in a dose-dependent manner (50-0.80 µM) for 48 (myoblasts) or 72 (myotubes) hours. Cell viability was tested using a 3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay and cell counting; myotube area was determined by immunocytochemistry and confocal microscopy; and anabolic signaling pathways were evaluated by western blot and RT-PCR. Results: Our data show that the treatment of primary human myoblasts treated with 50 µM and 25 µM of AA led to the release of PGE2 and PGF2α at levels higher than those of control-treated cells (p < 0.001 for all concentrations). Additionally, 50 µM and 25 µM of AA suppressed myoblast proliferation, myotube area, and myotube fusion. Anabolic signaling indicated reductions in total and phosphorylated TSC2, AKT, S6, and 4EBP1 in myoblasts at 50 µM of AA (p < 0.01 for all), but not in myotubes. These changes were not affected by COX-2 inhibition with celecoxib. Conclusion: Together, our data demonstrate that high concentrations of AA inhibit myoblast proliferation, myotube fusion, and myotube hypertrophy, thus revealing potential deleterious effects of AA on human skeletal muscle cell health and viability.


Asunto(s)
Fibras Musculares Esqueléticas , Mioblastos Esqueléticos , Humanos , Ratones , Animales , Ácido Araquidónico/farmacología , Diferenciación Celular , Hipertrofia/metabolismo , Músculo Esquelético
11.
FEBS Lett ; 597(9): 1225-1232, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36971014

RESUMEN

Macrophages play an important role in the response to infection and/or repair of injury in tissues. To examine the NF-κB pathway in response to an inflammatory stimulus, we used wild-type bone-marrow-derived macrophages (BMDMs) or BMDMs with knockout (KO) of myeloid differentiation primary response 88 (MyD88) and/or Toll/interleukin-1 receptor domain-containing adapter-inducing interferon-ß (TRIF) via CRISPR/Cas9. Following treatment of BMDMs with lipopolysaccharide (LPS) to induce an inflammatory response, translational signalling of NF-κB was quantified via immunoblot and cytokines were measured. Our findings reveal that MyD88 KO, but not TRIF KO, decreased LPS-induced NF-κB signalling, and 10% expression of basal MyD88 expression was sufficient to partially rescue the abolished inflammatory cytokine secretion observed upon MyD88 KO.


Asunto(s)
Lipopolisacáridos , FN-kappa B , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Lipopolisacáridos/toxicidad , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Animales , Ratones
12.
Growth Factors ; 30(6): 367-84, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23137199

RESUMEN

Phosphoinositide 3-kinase (PI3K) is a principal regulator of Akt activation and myogenesis; however, the function of PI3K p110ß in these processes is not well defined. To address this, we investigated the role of p110ß in Akt activation and skeletal muscle cell differentiation. We found that Akt phosphorylation was enhanced in p110ß-deficient myoblasts in response to Insulin-like Growth Factor-I (IGF-I), epidermal growth factor, or p110α overexpression, as compared to p110ß-sufficient cells. This effect was associated with increased mammalian target of rapamycin complex 2 activation, even in myoblasts deficient in mSin1 and rictor. Conversely, in response to the G-protein-coupled receptor agonist lysophosphatidic acid, Akt phosphorylation was attenuated in p110ß-deficient myoblasts. Loss of p110ß also enhanced the expression of myogenic markers at the myoblast stage and during the first 48 h of differentiation. These data demonstrate that reductions in p110ß are associated with agonist-specific Akt hyperactivation and accelerated myogenesis, thus revealing a negative role for p110ß in Akt activation and during myoblast differentiation.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Regulación de la Expresión Génica , Complejos Multiproteicos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Diferenciación Celular , Perfilación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lisofosfolípidos/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Fosforilación , ARN Interferente Pequeño/metabolismo , Regeneración , Transducción de Señal
13.
Physiol Rep ; 10(21): e15481, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36325583

RESUMEN

The use of non-steroidal anti-inflammatory drugs (NSAIDs) for treatment of musculoskeletal injuries is commonplace in the general, athletic, and military populations. While NSAIDs have been studied in a variety of tissues, the effects of NSAIDs on skeletal muscle have not been fully defined. To address this, we investigated the degree to which the cyclooxygenase (COX)-2-selective NSAID celecoxib affects muscle cell proliferation, differentiation, anabolic signaling, and mitochondrial function in primary human skeletal myoblasts and myotubes. Primary muscle cells were treated with celecoxib or NS-398 (a pharmacological inhibitor of COX-2) as a control. Celecoxib administration significantly reduced myoblast proliferation, viability, fusion, and myotube area in a dose-dependent manner, whereas NS-398 had no effect on any of these outcomes. Celecoxib treatment was also associated with reduced phosphorylation of ribosomal protein S6 in myoblasts, and reduced phosphorylation of AKT, p70S6K, S6, and ERK in myotubes. In contrast, NS-398 did not alter phosphorylation of these molecules in myoblasts or myotubes. In myoblasts, celecoxib significantly reduced mitochondrial membrane potential and respiration, as evidenced by the decreased citric acid cycle (CAC) intermediates cis-aconitic acid, alpha-keto-glutarate acid, succinate acid, and malic acid. Similar results were observed in myotubes, although celecoxib also reduced pyruvic acid, citric acid, and fumaric acid. NS-398 did not affect CAC intermediates in myoblasts or myotubes. Together, these data reveal that celecoxib inhibits proliferation, differentiation, intracellular signaling, and mitochondrial function in primary human myoblasts and myotubes independent of its function as a COX-2 inhibitor.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2 , Mioblastos Esqueléticos , Humanos , Celecoxib/farmacología , Ciclooxigenasa 2 , Diferenciación Celular/fisiología , Inhibidores de la Ciclooxigenasa 2/farmacología , Antiinflamatorios no Esteroideos/farmacología , Proliferación Celular
14.
J Appl Physiol (1985) ; 133(1): 170-182, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35678743

RESUMEN

This investigation examined the influence of 12-week ballistic resistance training programs on the IGF-I system in circulation, interstitial fluid, and skeletal muscle, at rest and in response to acute exercise. Seventeen college-aged subjects (11 women/6 men; 21.7 ± 3.7 yr) completed an acute ballistic exercise bout before and after the training program. Blood samples were collected pre-, mid-, and postexercise and analyzed for serum total IGF-I, free IGF-I, and IGF binding proteins (IGFBPs) 1-4. Dialysate and interstitial free IGF-I were analyzed in vastus lateralis (VL) interstitial fluid collected pre- and postexercise via microdialysis. Pre- and postexercise VL muscle biopsies were analyzed for IGF-I protein expression, IGF-I receptor phosphorylation (p-IGF-IR), and AKT phosphorylation (p-AKT). Following training, basal serum IGF-I, free IGF-I, IGFBP-2, and IGFBP-3 decreased whereas IGFBP-1 and IGFBP-4 increased. Training reduced basal dialysate and interstitial free IGF-I but had no effect on basal skeletal muscle IGF-I, p-IGF-IR, or p-AKT. Acute exercise elicited transient changes in IGF-I system concentrations and downstream anabolic signaling both pre- and posttraining; training did not affect this acute exercise response. Posttraining, acute exercise-induced changes in dialysate/interstitial free IGF-I were strongly correlated with the changes in intramuscular IGF-I expression, p-IGF-IR, and p-AKT. The divergent influence of resistance training on circulating/interstitial and skeletal muscle IGF-I demonstrates the importance of concurrent, multiple biocompartment analysis when examining the IGF-I system. As training elicited muscle hypertrophy, these findings indicate that IGF-I's anabolic effects on skeletal muscle are mediated by local, rather than systemic mechanisms.NEW & NOTEWORTHY In the first investigation to assess resistance training's effects on the IGF-I system in serum, interstitial fluid, and skeletal muscle, training decreased basal circulating and interstitial IGF-I but did not alter basal intramuscular IGF-I protein activity. Posttraining, acute exercise-induced interstitial IGF-I increases were strongly correlated with intramuscular IGF-I expression and signaling. These findings highlight the importance of multibiocompartment measurement when analyzing IGF-I and suggest that IGF-I's role in hypertrophic adaptations is locally mediated.


Asunto(s)
Ejercicio Físico , Líquido Extracelular , Factor I del Crecimiento Similar a la Insulina , Entrenamiento de Fuerza , Ejercicio Físico/fisiología , Líquido Extracelular/metabolismo , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Músculo Esquelético/fisiología , Proteínas Proto-Oncogénicas c-akt , Adulto Joven
15.
J Biol Methods ; 8(3): e151, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34514012

RESUMEN

Genomic manipulation offers the possibility for novel therapies in lieu of medical interventions in use today. The ability to genetically restore missing inflammatory genes will have a monumental impact on our current immunotherapy treatments. This study compared the efficacy of two different genetic manipulation techniques: clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) transfection to adenoviral transduction to determine which method would provide the most transient and stable knockdown of myeloid differentiation primary response 88 (MyD88). MyD88 is a major regulator of nuclear factor kappa light chain enhancer of activated B cells (NFκB) pathway in Raw 264.7 macrophages. Following genetic manipulation, cells were treated for 24 h with Lipopolysaccharide (LPS) to stimulate the inflammatory pathway. Confirmation of knockdown was determined by western immunoblotting and quantification of band density. Both CRISPR/Cas9 and adenoviral transduction produced similar knockdown efficiency (~64% and 60%, respectively) in MyD88 protein 48 h post adenoviral transduction. NFκB phosphorylation was increased in CRISPR/Cas9-mediated MyD88 knockdown and control cells, but not in adenovirus-mediated MyD88 knockdown cells, following LPS administration. CRISPR/Cas9-mediated MyD88 knockdown macrophages treated with LPS for 24 h showed a 65% reduction in tumor necrosis factor alpha (TNFα) secretion, and a 67% reduction in interleukin-10 (IL-10) secretion when compared to LPS-stimulated control cells (P ≤ 0.01 for both). LPS did not stimulate TNFα or IL-10 secretion in adenovirus-mediated control or MyD88 knockdown cells. These data demonstrate that Raw 264.7 macrophages maintain responsiveness to inflammatory stimuli following CRISPR/Cas9-mediated reductions in MyD88, but not following adenovirus-mediated MyD88 knockdown.

16.
Exp Biol Med (Maywood) ; 246(9): 1104-1111, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33641442

RESUMEN

There is mounting evidence suggesting that the commonly used analgesics, non-steroidal anti-inflammatory drugs (NSAIDs), may inhibit new bone formation with physical training and increase risk of stress fractures in physically active populations. Stress fractures are thought to occur when bones are subjected to repetitive mechanical loading, which can lead to a cycle of tissue microdamage, repair, and continued mechanical loading until fracture. Adaptive bone formation, particularly on the periosteal surface of long bones, is a concurrent adaptive response of bone to heightened mechanical loading that can improve the fatigue resistance of the skeletal structure, and therefore may play a critical role in offsetting the risk of stress fracture. Reports from animal studies suggest that NSAID administration may suppress this important adaptive response to mechanical loading. These observations have implications for populations such as endurance athletes and military recruits who are at risk of stress fracture and whose use of NSAIDs is widespread. However, results from human trials evaluating exercise and bone adaptation with NSAID consumption have been less conclusive. In this review, we identify knowledge gaps that must be addressed to further support NSAID-related guidelines intended for at-risk populations and individuals.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Remodelación Ósea/efectos de los fármacos , Fracturas por Estrés , Osteogénesis/efectos de los fármacos , Animales , Fracturas por Estrés/inducido químicamente , Fracturas por Estrés/fisiopatología , Humanos
17.
Growth Factors ; 28(5): 318-28, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20380591

RESUMEN

We previously showed that exogenous insulin-like growth factor-I (IGF-I) and bone morphogenetic protein-7 (BMP-7) synergistically stimulated osteoblast differentiation in fetal rat calvaria (FRC) cells. We have now shown that BMP-7 alone and the BMP-7 and IGF-I combination synergistically stimulated protein kinase D (PKD) phosphorylation at Ser744/748 and Ser916. Transfection of FRC cells with a constitutively active PKD stimulated marker expression, while transfection with a catalytically inactive PKD did not. Moreover, Gö6976, which inhibits protein kinase C (PKC) α and β1, blocked PKD phosphorylation and the synergistic action of the BMP-7 and IGF-I combination on osteoblast differentiation, whereas Gö6983, which inhibits PKCα, β, γ, δ, and ζ, did not. Our results suggest that the FRC cell differentiation induced by BMP-7 and the BMP-7 and IGF-I combination requires stimulation of PKD activity. Our results are consistent with a novel mechanism in which combined BMP-7 and IGF-I signaling activates upstream novel PKC(s), which then phosphorylates and activates PKD, leading to enhanced osteoblast differentiation.


Asunto(s)
Proteína Morfogenética Ósea 7/farmacología , Diferenciación Celular/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Osteoblastos/efectos de los fármacos , Proteína Quinasa C/metabolismo , Animales , Carbazoles/farmacología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Osteoblastos/enzimología , Osteoblastos/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C beta , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/metabolismo , Ratas
18.
Biochem Biophys Res Commun ; 390(2): 252-7, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19799871

RESUMEN

The PI3K/Akt/mTOR signaling pathway is critical for cellular growth and survival in skeletal muscle, and is activated in response to growth factors such as insulin-like growth factor-I (IGF-I). We found that in C2C12 myoblasts, deficiency of PI3K p110 catalytic subunits or Akt isoforms had distinct effects on phosphorylation of mTOR and p70S6K. siRNA-mediated knockdown of PI3K p110alpha, p110beta, and simultaneous knockdown of p110alpha and p110beta resulted in increased basal and IGF-I-stimulated phosphorylation of mTOR S2448 and p70S6K T389; however, phosphorylation of S6 was reduced in p110beta-deficient cells, possibly due to reductions in total S6 protein. We found that IGF-I-stimulated Akt1 activity was enhanced in Akt2- or Akt3-deficient cells, and that knockdown of individual Akt isoforms increased mTOR/p70S6K activation in an isoform-specific fashion. Conversely, levels of IGF-I-stimulated p70S6K phosphorylation in cells simultaneously deficient in both Akt1 and Akt3 were increased beyond those seen with loss of any single Akt isoform, suggesting an alternate, Akt-independent mechanism that activates mTOR/p70S6K. Our results collectively suggest that mTOR/p70S6K is activated in a PI3K/Akt-dependent manner, but that in the absence of p110alpha or Akt, alternate pathway(s) may mediate activation of mTOR/p70S6K in C2C12 myoblasts.


Asunto(s)
Proteínas Portadoras/metabolismo , Mioblastos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Animales , Dominio Catalítico , Activación Enzimática , Técnicas de Silenciamiento del Gen , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones , Mioblastos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR
19.
Biochem Biophys Res Commun ; 389(1): 117-21, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19703413

RESUMEN

Oxidative stress has been shown to induce apoptosis in a variety of tissues, while insulin-like growth factor-I (IGF-I) can oppose this effect. We found that H(2)O(2) promoted cell death and apoptosis in C2C12 myoblasts, an effect that was completely prevented by exogenous IGF-I. One downstream mediator of IGF-I survival signaling is the serine/threonine kinase Akt, of which three isoforms have been identified in mammals. We found that Akt1 and Akt3 act on pro-apoptotic target molecules in an isoform-specific manner. Both Akt1 and Akt3 were responsible for phosphorylating FoxO3a at S253 and FoxO1 at T24, while Akt1 alone phosphorylated Bad at S136 and FoxO3a at T32. Our results provide evidence for IGF-I-stimulated isoform-specific actions of Akt on molecules involved in promoting apoptosis.


Asunto(s)
Apoptosis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Mioblastos/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular , Supervivencia Celular , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Peróxido de Hidrógeno/toxicidad , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Mioblastos/enzimología , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-akt/genética , ARN Interferente Pequeño/genética , Transducción de Señal , Proteína Letal Asociada a bcl/metabolismo
20.
Exp Biol Med (Maywood) ; 234(2): 164-70, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19064939

RESUMEN

The involvement of circulating insulin-like growth factor-I (IGF-I) in the skeletal muscle response to resistance exercise is currently unclear. To address this, we utilized the liver IGF-I-deficient (LID) mouse model, in which the igf1 gene has been disrupted in the hepatocytes, resulting in ~80% reduction in serum IGF-I. Twelve- to 13-month-old male LID and control (L/L) mice were subjected to 16 weeks of resistance training. Resistance exercise resulted in equal strength gains in both L/L and LID mice. Basal IGF-I mRNA levels were greater in LID muscles than in L/L, and exercise increased IGF-I mRNA in quadriceps, gastrocnemius, and plantaris muscles. LID mice had elevated tyrosine phosphorylation of IGF-IR and Stat5b, the latter possibly reflective of increased serum GH. Tyrosine phosphorylation of IGF-IR was increased, while phospho-Stat5b was reduced after resistance training of both wild-type and LID mice. These data suggest that: 1) performance and recovery in response to resistance training is normal even when there is severe deficiency of circulating IGF-I; and 2) upregulation of local IGF-I may be involved in the compensatory growth of muscle that occurs in response to resistance training. Decreased levels of p-Stat5b in exercised mice suggests that the upregulation of local IGF-I gene expression in response to exercise may be GH-independent.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/deficiencia , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Condicionamiento Físico Animal/fisiología , Animales , Peso Corporal , Regulación de la Expresión Génica , Hipertrofia , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Tamaño de los Órganos , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda