Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Undersea Hyperb Med ; 51(1): 85-92, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38615357

RESUMEN

This study aimed to investigate what factors determine freedivers' maximal static apnea dive time. We correlated some physical/physiological factors with male freedivers' maximum apnea diving duration. Thirty-six experienced male freedivers participated in this study. The divers participated in two days of the experiments. On the first day, apnea diving time, blood oxygen saturation (SpO2), heart rate (HR), blood pressure (BP), stress index, and blood parameters were measured before, during, and after the apnea diving in the pool. On the second day, body composition, lung capacity, resting and maximal oxygen consumption (VO2max), and the Wingate anaerobic power were measured in the laboratory. The data were analyzed with Pearson's Correlation using the SPSS 22 program. The correlation coefficient (R) of determination was set at 0.4, and the level of significance was set at p <0.05. There were positive correlations of diving experience, maximum SpO2, and lung capacity with the maximum apnea time R>0.4, P<0.05). There were negative correlations of BMI, body fat percentage, body fat mass, minimum SpO2, stress index, and total cholesterol with the maximum apnea diving time (R>-0.4, P<0.05). No correlations of age, height, weight, fat-free mass, skeletal muscle mass, HR, BP, blood glucose, beta- hydroxybutyrate, lactate, and hemoglobin levels with the maximum apnea diving time were observed (R<0.4, P>0.05). It is concluded that more experience in freediving, reduced body fat, extended SpO2 range, and increased lung capacity are the performance predictors and beneficial for freedivers to improve their maximum apnea diving performance.


Asunto(s)
Apnea , Buceo , Humanos , Apnea/etiología , Ácido 3-Hidroxibutírico , Glucemia , Ácido Láctico
2.
Prostate Int ; 8(2): 70-77, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32647643

RESUMEN

BACKGROUND: Recent studies have suggested a novel therapeutic strategy for treatment of benign prostatic hyperplasia (BPH) via modulation of autophagy. However, it is not clear whether autophagy induction or inhibition can render better therapeutic efficacy for BPH treatment because autophagy activation in BPH tissue is not precisely known and still contradictory. The purpose of this study was to examine the levels of autophagy in BPH tissue cells. METHODS: We have analyzed and compared autophagic flux which is defined as a measure of autophagic degradation activity in two human prostate epithelial cell lines, RWPE-1 (normal prostate) and BPH-1 (BPH) using LC3-II turnover assay, to clarify the levels of autophagy in BPH. RESULTS: The in vitro autophagy flux assays showed that autophagy flux was significantly decreased in BPH-1 cell lines compared with RWPE-1 cells under all three conditions of using the original (~62%), the exchanged (~46%), and the same media (Hank's balanced salt solution (HBSS), ~40%), and these results were similar to those seen in the prostate of testosterone-induced BPH rats (~50%) (P < 0.05). CONCLUSION: It is suggested that defective autophagy, which is decreased autophagy flux in the prostate gland, may be implicated in BPH, and activating autophagy flux of the prostate with BPH may be used as a potential therapeutic target for treating and alleviating BPH disease.

3.
Undersea Hyperb Med ; 47(1): 93-100, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32176950

RESUMEN

The purpose of this study was to investigate the effects of a single bout of heliox non-saturation diving on the cardiovascular system and cognitive function. Ten recreational scuba divers (10 males, ∼35 years old) participated in this study. These subjects made two pool dives within a one-week interval, alternating gases with compressed air (21% O2, 79% N2) and with heliox (21% O2 and 79% He). The depth was to 26 meters over a 20-minute duration. The results showed that heliox diving significantly increased blood O2 saturation by 1.15% and significantly decreased blood lactate levels by ∼57% when compared with air diving (P ≺ 0.05). However, there were no significant differences in resting heart rate, systolic or diastolic pressure, core body blood pressure, and pulse wave velocity between the heliox and air dives. The Stroop test showed that the heliox dive significantly increased cognitive function compared with the air dive in both the simple test (Offtime) and interference test (Ontime) (P ≺ 0.05). It was concluded that the heliox dive increases blood O2 saturation and decreases blood lactate concentration when compared with air dives. These conditions are likely to help divers reduce hypoxia in the water, reduce the risk of loss of consciousness, reduce fatigue and allow them to dive for longer. Heliox diving may also help judgment and risk coping skills in the water due to the improvement of cognitive ability as compared to air breathing dives.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Cognición/efectos de los fármacos , Buceo/fisiología , Helio/farmacología , Ácido Láctico/sangre , Oxígeno/sangre , Adulto , Aire , Presión Sanguínea/efectos de los fármacos , Hipoxia de la Célula , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Hipoxia/sangre , Hipoxia/prevención & control , Masculino , Oxígeno/farmacología , Análisis de la Onda del Pulso , Recreación , Factores de Tiempo , Rigidez Vascular
4.
Data Brief ; 13: 696-699, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28725675

RESUMEN

We analyzed autophagy/mitophagy flux in vitro (C2C12 myotubes) and in vivo (mouse skeletal muscle) following the treatments of autophagy inducers (starvation, rapamycin) and a mitophagy inducer (carbonyl cyanide m-chlorophenylhydrazone, CCCP) using two immunodetection methods, ELISA and Western blotting, and compared their working range, accuracy, and reliability. The ELISAs showed a broader working range than that of the LC3 Western blots (Table 1). Table 2 showed that data value distribution was tighter and the average standard error from the ELISA was much smaller than those of the Western blot, directly relating to the accuracy of the assay. Test-retest reliability analysis showed good reliability for three individual ELISAs (interclass correlation, ≥ 0.7), but poor reliability for three individual Western blots (interclass correlation, ≤ 0.4) (Table 3).

5.
Anal Biochem ; 530: 57-67, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28477964

RESUMEN

Macroautophagy (hereafter referred to as autophagy) is a degradation system that delivers cytoplasmic materials to lysosomes via autophagosomes. Autophagic flux is defined as a measure of autophagic degradation activity. Despite several methods for monitoring autophagic flux being currently utilized, interest in finding a highly accurate, sensitive and well-quantifiable assay is still growing. Therefore, we introduce a new approach analyzing autophagic flux in vitro and in vivo using enzyme-linked immunosorbent assay (ELISA) technique. In order to adapt this assay from LC3-II turnover measured by Western blot in the presence and absence of lysosomal inhibitors, we induced autophagy by starvation or rapamycin and mitophagy (mitochondrial degradation by autophagy) by CCCP in C2C12 myotubes for 8 h and in mice for 48 h with and without Bafilomycin A1 or colchicine treatment, respectively. Following subcellular fractionation of mouse skeletal muscle cells and tissue, cytosolic, membrane, and mitochondrial fractions were analyzed through a sandwich ELISA using two LC3 antibodies, LC3 capture and HRP-conjugated LC3 detection antibodies. Using this ELISA, changes in the membrane-bound or mitochondrion-associated LC3-II levels, and the ratio of the LC3-II from each fraction to LC3-I levels (cytosolic fraction) were evaluated for measuring autophagy and mitophagy flux. This study demonstrates that this ELISA was more sensitive and reliable to measure autophagic/mitophagic flux in both in vitro and in vivo, compared with the most commonly used LC3 turnover assay via Western blot.


Asunto(s)
Autofagia , Ensayo de Inmunoadsorción Enzimática/métodos , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/patología , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Animales , Western Blotting , Compartimento Celular , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo
6.
J Physiol Sci ; 66(5): 417-30, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26943341

RESUMEN

Mitochondrial homeostasis is tightly regulated by two major processes: mitochondrial biogenesis and mitochondrial degradation by autophagy (mitophagy). Research in mitochondrial biogenesis in skeletal muscle in response to endurance exercise training has been well established, while the mechanisms regulating mitophagy and the interplay between mitochondrial biogenesis and degradation following endurance exercise training are not yet well defined. The purpose of this study was to examine the effects of a short-term inhibition of autophagy in response to acute endurance exercise on skeletal muscle mitochondrial biogenesis and dynamics in an exercise-trained condition. Male wild-type C57BL/6 mice performed five daily bouts of 1-h swimming per week for 8 weeks. In order to measure autophagy flux in mouse skeletal muscle, mice were treated with or without 2 days of 0.4 mg/kg/day intraperitoneal colchicine (blocking the degradation of autophagosomes) following swimming exercise training. The autophagic flux assay demonstrated that swimming training resulted in an increase in the autophagic flux (~100 % increase in LC3-II) in mouse skeletal muscle. Mitochondrial fusion proteins, Opa1 and MFN2, were significantly elevated, and mitochondrial fission protein, Drp1, was also increased in trained mouse skeletal muscle, suggesting that endurance exercise training promotes both mitochondrial fusion and fission processes. A mitochondrial receptor, Bnip3, was further increased in exercised muscle when treated with colchicine while Pink/Parkin protein levels were unchanged. The endurance exercise training induced increases in mitochondrial biogenesis marker proteins, SDH, COX IV, and a mitochondrial biogenesis promoting factor, PGC-1α but this effect was abolished in colchicine-treated mouse skeletal muscle. This suggests that autophagy plays an important role in mitochondrial biogenesis and this coordination between these opposing processes is involved in the cellular adaptation to endurance exercise training.


Asunto(s)
Autofagia/fisiología , Músculo Esquelético/metabolismo , Biogénesis de Organelos , Condicionamiento Físico Animal/fisiología , Resistencia Física/fisiología , Animales , Colchicina/farmacología , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Natación/fisiología
7.
Autophagy ; 9(12): 2115-25, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24184927

RESUMEN

Colchicine treatment is associated with an autophagic vacuolar myopathy in human patients. The presumed mechanism of colchicine-induced myotoxicity is the destabilization of the microtubule system that leads to impaired autophagosome-lysosome fusion and the accumulation of autophagic vacuoles. Using the MTOR inhibitor rapamycin we augmented colchicine's myotoxic effect by increasing the autophagic flux; this resulted in an acute myopathy with muscle necrosis. In contrast to myonecrosis induced by cardiotoxin, myonecrosis induced by a combination of rapamycin and colchicine was associated with accumulation of autophagic substrates such as LC3-II and SQSTM1; as a result, autophagic vacuoles accumulated in the center of myofibers, where LC3-positive autophagosomes failed to colocalize with the lysosomal protein marker LAMP2. A similar pattern of central LC3 accumulation and myonecrosis is seen in human patients with colchicine myopathy, many of whom have been treated with statins (HMGCR/HMG-CoA reductase inhibitors) in addition to colchicine. In mice, cotreatment with colchicine and simvastatin also led to muscle necrosis and LC3 accumulation, suggesting that, like rapamycin, simvastatin activates autophagy. Consistent with this, treatment of mice with four different statin medications enhanced autophagic flux in skeletal muscle in vivo. Polypharmacy is a known risk factor for toxic myopathies; our data suggest that some medication combinations may simultaneously activate upstream autophagy signaling pathways while inhibiting the degradation of these newly synthesized autophagosomes, resulting in myotoxicity.


Asunto(s)
Autofagia/fisiología , Colchicina/farmacología , Enfermedades por Almacenamiento Lisosomal/inducido químicamente , Enfermedades Musculares/inducido químicamente , Animales , Autofagia/efectos de los fármacos , Progresión de la Enfermedad , Humanos , Enfermedades por Almacenamiento Lisosomal/patología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/patología , Fagosomas/efectos de los fármacos , Fagosomas/patología , Simvastatina/farmacología , Regulación hacia Arriba/efectos de los fármacos , Vacuolas/efectos de los fármacos , Vacuolas/patología
8.
Hum Mol Genet ; 22(6): 1167-79, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23250913

RESUMEN

Autophagy is dysfunctional in many degenerative diseases including myopathies. Mutations in valosin-containing protein (VCP) cause inclusion body myopathy (IBM) associated with Paget's disease of the bone, fronto-temporal dementia and amyotrophic lateral sclerosis (IBMPFD/ALS). VCP is necessary for protein degradation via the proteasome and lysosome. IBMPFD/ALS mutations in VCP disrupt autophagosome and endosome maturation resulting in vacuolation, weakness and muscle atrophy. To understand the regulation of autophagy in VCP-IBM muscle, we examined the AKT/FOXO3 and mammalian target of rapamycin (mTOR) pathways. Basal Akt and FOXO3 phosphorylation was normal. In contrast, the phosphorylation of mTOR targets was decreased. Consistent with this, global protein translation was diminished and autophagosome biogenesis was increased in VCP-IBM muscle. Further mTORC1 inhibition with rapamycin hastened weakness, atrophy and vacuolation in VCP-IBM mice. This was accompanied by the accumulation of autophagic substrates such as p62, LC3II and ubiquitinated proteins. The decrease in mTOR signaling was partially rescued by insulin and to a lesser extent by amino acid (AA) stimulation in VCP-IBM muscle. Cells expressing catalytically inactive VCP or treated with a VCP inhibitor also failed to activate mTOR upon nutrient stimulation. Expression of a constitutively active Rheb enhanced mTOR activity and increased the fiber size in VCP-IBM mouse skeletal muscle. These studies suggest that VCP mutations may disrupt mTOR signaling and contribute to IBMPFD/ALS disease pathogenesis. Treatment of some autophagic disorders with mTOR inhibitors such as rapamycin may worsen disease.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Miositis por Cuerpos de Inclusión/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Vacuolas/metabolismo , Adenosina Trifosfatasas/genética , Animales , Autofagia , Proteínas de Ciclo Celular/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miositis por Cuerpos de Inclusión/genética , Miositis por Cuerpos de Inclusión/patología , Miositis por Cuerpos de Inclusión/fisiopatología , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Vacuolas/patología , Proteína que Contiene Valosina
9.
Mol Cell Biol ; 32(14): 2837-48, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22586267

RESUMEN

AMP activated protein kinase (AMPK) plays a key role in the regulatory network responsible for maintaining systemic energy homeostasis during exercise or nutrient deprivation. To understand the function of the regulatory ß2 subunit of AMPK in systemic energy metabolism, we characterized ß2 subunit-deficient mice. Using these mutant mice, we demonstrated that the ß2 subunit plays an important role in regulating glucose, glycogen, and lipid metabolism during metabolic stress. The ß2 mutant animals failed to maintain euglycemia and muscle ATP levels during fasting. In addition, ß2-deficient animals showed classic symptoms of metabolic syndrome, including hyperglycemia, glucose intolerance, and insulin resistance when maintained on a high-fat diet (HFD), and were unable to maintain muscle ATP levels during exercise. Cell surface-associated glucose transporter levels were reduced in skeletal muscle from ß2 mutant animals on an HFD. In addition, they displayed poor exercise performance and impaired muscle glycogen metabolism. These mutant mice had decreased activation of AMPK and deficits in PGC1α-mediated transcription in skeletal muscle. Our results highlight specific roles of AMPK complexes containing the ß2 subunit and suggest the potential utility of AMPK isoform-specific pharmacological modulators for treatment of metabolic, cardiac, and neurological disorders.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Ayuno/metabolismo , Glucosa/metabolismo , Homeostasis , Redes y Vías Metabólicas , Síndrome Metabólico/etiología , Síndrome Metabólico/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Esfuerzo Físico/fisiología , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ribonucleótidos/farmacología , Estrés Fisiológico , Distribución Tisular
10.
Autophagy ; 6(7): 929-35, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20657169

RESUMEN

Reliable and quantitative assays to measure in vivo autophagy are essential. Currently, there are varied methods for monitoring autophagy; however, it is a challenge to measure "autophagic flux" in an in vivo model system. Conversion and subsequent degradation of the microtubule-associated protein 1 light chain 3 (MAP1-LC3/LC3) to the autophagosome associated LC3-II isoform can be evaluated by immunoblot. However, static levels of endogenous LC3-II protein may render possible misinterpretations since LC3-II levels can increase, decrease or remain unchanged in the setting of autophagic induction. Therefore, it is necessary to measure LC3-II protein levels in the presence and absence of lysomotropic agents that block the degradation of LC3-II, a technique aptly named the "autophagometer." In order to measure autophagic flux in mouse skeletal muscle, we treated animals with the microtubule depolarizing agent colchicine. Two days of 0.4 mg/kg/day intraperitoneal colchicine blocked autophagosome maturation to autolysosomes and increased LC3-II protein levels in mouse skeletal muscle by >100%. The addition of an autophagic stimulus such as dietary restriction or rapamycin led to an additional increase in LC3-II above that seen with colchicine alone. Moreover, this increase was not apparent in the absence of a "colchicine block." Using this assay, we evaluated the autophagic response in skeletal muscle upon denervation induced atrophy. Our studies highlight the feasibility of performing an "in vivo autophagometer" study using colchicine in skeletal muscle.


Asunto(s)
Autofagia/fisiología , Músculo Esquelético/fisiología , Animales , Antirreumáticos/farmacología , Línea Celular , Cloroquina/farmacología , Colchicina/farmacología , Humanos , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Desnervación Muscular , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Moduladores de Tubulina/farmacología , Vinblastina/farmacología
11.
Hum Mol Genet ; 19(R1): R38-45, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20410287

RESUMEN

Inclusion body myopathy associated with Paget's disease of the bone and fronto-temporal dementia (IBMPFD) is a progressive autosomal dominant disorder caused by mutations in p97/VCP (valosin-containing protein). p97/VCP is a member of the AAA+ (ATPase associated with a variety of activities) protein family and participates in multiple cellular processes. One particularly important role for p97/VCP is facilitating intracellular protein degradation. p97/VCP has traditionally been thought to mediate the ubiquitin-proteasome degradation of proteins; however, recent studies challenge this dogma. p97/VCP clearly participates in the degradation of aggregate-prone proteins, a process principally mediated by autophagy. In addition, IBMPFD mutations in p97/VCP lead to accumulation of autophagic structures in patient and transgenic animal tissue. This is likely due to a defect in p97/VCP-mediated autophagosome maturation. The following review will discuss the evidence for p97/VCP in autophagy and how a disruption in this process contributes to IBMPFD pathogenesis.


Asunto(s)
Autofagia , Demencia Frontotemporal/complicaciones , Miositis por Cuerpos de Inclusión/complicaciones , Osteítis Deformante/complicaciones , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Demencia Frontotemporal/genética , Homeostasis , Humanos , Mutación , Miositis por Cuerpos de Inclusión/genética , Osteítis Deformante/genética , Proteína que Contiene Valosina
12.
Hum Mol Genet ; 19(7): 1165-73, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20045868

RESUMEN

Distal arthrogryposis type I (DA1) is a disorder characterized by congenital contractures of the hands and feet for which few genes have been identified. Here we describe a five-generation family with DA1 segregating as an autosomal dominant disorder with complete penetrance. Genome-wide linkage analysis using Affymetrix GeneChip Mapping 10K data from 12 affected members of this family revealed a multipoint LOD(max) of 3.27 on chromosome 12q. Sequencing of the slow-twitch skeletal muscle myosin binding protein C1 (MYBPC1), located within the linkage interval, revealed a missense mutation (c.706T>C) that segregated with disease in this family and causes a W236R amino acid substitution. A second MYBPC1 missense mutation was identified (c.2566T>C)(Y856H) in another family with DA1, accounting for an MYBPC1 mutation frequency of 13% (two of 15). Skeletal muscle biopsies from affected patients showed type I (slow-twitch) fibers were smaller than type II fibers. Expression of a green fluorescent protein (GFP)-tagged MYBPC1 construct containing WT and DA1 mutations in mouse skeletal muscle revealed robust sarcomeric localization. In contrast, a more diffuse localization was seen when non-fused GFP and MYBPC1 proteins containing corresponding MYBPC3 amino acid substitutions (R326Q, E334K) that cause hypertrophic cardiomyopathy were expressed. These findings reveal that the MYBPC1 is a novel gene responsible for DA1, though the mechanism of disease may differ from how some cardiac MYBPC3 mutations cause hypertrophic cardiomyopathy.


Asunto(s)
Artrogriposis/genética , Proteínas Portadoras/genética , Artrogriposis/patología , Secuencia de Bases , Femenino , Genes Dominantes , Humanos , Masculino , Datos de Secuencia Molecular , Mutación Missense
14.
J Cell Biol ; 187(6): 875-88, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-20008565

RESUMEN

Mutations in valosin-containing protein (VCP) cause inclusion body myopathy (IBM), Paget's disease of the bone, and frontotemporal dementia (IBMPFD). Patient muscle has degenerating fibers, rimmed vacuoles (RVs), and sarcoplasmic inclusions containing ubiquitin and TDP-43 (TARDNA-binding protein 43). In this study, we find that IBMPFD muscle also accumulates autophagosome-associated proteins, Map1-LC3 (LC3), and p62/sequestosome, which localize to RVs. To test whether VCP participates in autophagy, we silenced VCP or expressed adenosine triphosphatase-inactive VCP. Under basal conditions, loss of VCP activity results in autophagosome accumulation. After autophagic induction, these autophagosomes fail to mature into autolysosomes and degrade LC3. Similarly, IBMPFD mutant VCP expression in cells and animals leads to the accumulation of nondegradative autophagosomes that coalesce at RVs and fail to degrade aggregated proteins. Interestingly, TDP-43 accumulates in the cytosol upon autophagic inhibition, similar to that seen after IBMPFD mutant expression. These data implicate VCP in autophagy and suggest that impaired autophagy explains the pathology seen in IBMPFD muscle, including TDP-43 accumulation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Autofagia , Proteínas de Ciclo Celular/metabolismo , Demencia Frontotemporal/enzimología , Miositis por Cuerpos de Inclusión/enzimología , Osteítis Deformante/enzimología , Músculo Cuádriceps/enzimología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas/genética , Animales , Autofagia/genética , Biopsia , Estudios de Casos y Controles , Proteínas de Ciclo Celular/genética , Línea Celular , Cloroquina , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Demencia Frontotemporal/inducido químicamente , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Proteínas de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Miositis por Cuerpos de Inclusión/inducido químicamente , Miositis por Cuerpos de Inclusión/genética , Miositis por Cuerpos de Inclusión/patología , Osteítis Deformante/inducido químicamente , Osteítis Deformante/genética , Osteítis Deformante/patología , Músculo Cuádriceps/patología , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Proteína Sequestosoma-1 , Transfección , Ubiquitina/metabolismo , Proteína que Contiene Valosina
15.
Autophagy ; 5(4): 511-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19305149

RESUMEN

The analysis of autophagy in cells and tissue has principally been performed via qualitative measures. These assays identify autophagosomes or measure the conversion of LC3I to LC3II. However, qualitative assays fail to quantitate the degradation of an autophagic substrate and therefore only indirectly measure an intact autophagic system. "Autophagic flux" can be measured using long-lived proteins that are degraded via autophagy. We developed a quantifiable luciferase reporter assay that measures the degradation of a long-lived polyglutamine protein aggregate, polyQ80-luciferase. Using this reporter, the induction of autophagy via starvation or rapamycin in cells preferentially decreases polyQ80-luciferase when compared with a nonaggregating polyQ19-luciferase after four hours of treatment. This response was both time- and concentration-dependent, prevented by autophagy inhibitors and absent in ATG5 knockout cells. We adapted this assay to living animals by electroporating polyQ19-luciferase and polyQ80-luciferase expression constructs into the right and left tibialis anterior (TA) muscles of mice, respectively. The change in the ratio of polyQ80-luciferase to polyQ19-luciferase signal before and after autophagic stimulation or inhibition was quantified via in vivo bioluminescent imaging. Following two days of starvation or treatment with intraperitoneal rapamycin, there was a approximately 35% reduction in the ratio of polyQ80:polyQ19-luciferase activity, consistent with the selective autophagic degradation of polyQ80 protein. This autophagic response in skeletal muscle in vivo was abrogated by co-treatment with chloroquine and in ATG16L1 hypomorphic mice. Our study demonstrates a method to quantify the autophagic flux of an expanded polyglutamine via luciferase reporters in vitro and in vivo.


Asunto(s)
Autofagia , Bioensayo/métodos , Genes Reporteros , Luciferasas/metabolismo , Péptidos/química , Péptidos/metabolismo , Animales , Autofagia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Estructura Cuaternaria de Proteína , Sirolimus/farmacología , Extractos de Tejidos , Transfección
16.
J Biol Chem ; 283(44): 30289-99, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18715868

RESUMEN

Mutations in p97/VCP cause the multisystem disease inclusion body myopathy, Paget disease of the bone and frontotemporal dementia (IBMPFD). p97/VCP is a member of the AAA+ (ATPase associated with a variety of activities) protein family and has been implicated in multiple cellular processes. One pathologic feature in IBMPFD is ubiquitinated inclusions, suggesting that mutations in p97/VCP may affect protein degradation. The present study shows that IBMPFD mutant expression increases ubiquitinated proteins and susceptibility to proteasome inhibition. Co-expression of an aggregate prone protein such as expanded polyglutamine in IBMPFD mutant cells results in an increase in aggregated protein that localizes to small inclusions instead of a single perinuclear aggresome. These small inclusions fail to co-localize with autophagic machinery. IBMPFD mutants avidly bind to these small inclusions and may not allow them to traffic to an aggresome. This is rescued by HDAC6, a p97/VCP-binding protein that facilitates the autophagic degradation of protein aggregates. Expression of HDAC6 improves aggresome formation and protects IBMPFD mutant cells from polyglutamine-induced cell death. Our study emphasizes the importance of protein aggregate trafficking to inclusion bodies in degenerative diseases and the therapeutic benefit of inclusion body formation.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Proteínas de Ciclo Celular/fisiología , Adenosina Trifosfatasas/metabolismo , Animales , Autofagia , Proteínas de Ciclo Celular/metabolismo , Muerte Celular , Línea Celular Tumoral , Femenino , Humanos , Cuerpos de Inclusión , Ratones , Modelos Biológicos , Mutación , Péptidos/química , Plásmidos/metabolismo , Unión Proteica , Ubiquitina/química , Proteína que Contiene Valosina
17.
Am J Physiol Cell Physiol ; 292(1): C564-72, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16870829

RESUMEN

Data from the use of activators and inhibitors of the AMP-activated protein kinase (AMPK) suggest that AMPK increases sensitivity of glucose transport to stimulation by insulin in muscle cells. We assayed insulin action after adenoviral (Ad) transduction of constitutively active (CA; a truncated form of AMPKalpha(1)) and dominant-negative (DN; which depletes endogenous AMPKalpha) forms of AMPKalpha (Ad-AMPKalpha-CA and Ad-AMPKalpha-DN, respectively) into C(2)C(12) myotubes. Compared with control (Ad-green fluorescent protein), Ad-AMPK-CA increased the ability of insulin to stimulate glucose transport. The increased insulin action in cells expressing AMPK-CA was suppressed by compound C (an AMPK inhibitor). Exposure of cells to 5-aminoimidazole-4-carboxamide-1beta-D-ribofuranoside (an AMPK activator) increased insulin action in uninfected myotubes and myotubes transduced with green fluorescent protein but not in Ad-AMPK-DN-infected myotubes. In Ad-AMPK-CA-transduced cells, serine phosphorylation of insulin receptor substrate 1 was decreased at a mammalian target of rapamycin (or p70 S6 kinase) target site that has been reported to be associated with insulin resistance. These data suggest that, in myotubes, activated AMPKalpha(1) is sufficient to increase insulin action and that the presence of functional AMPKalpha is required for 5-aminoimidazole-4-carboxamide-1beta,D-ribofuranoside-related increases in insulin action.


Asunto(s)
Glucosa/metabolismo , Hipoglucemiantes/metabolismo , Insulina/metabolismo , Complejos Multienzimáticos/farmacología , Proteínas Serina-Treonina Quinasas/farmacología , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Activadores de Enzimas/farmacología , Genes Dominantes , Proteínas Fluorescentes Verdes , Proteínas Sustrato del Receptor de Insulina , Isoenzimas/farmacología , Sustancias Luminiscentes , Ratones , Complejos Multienzimáticos/genética , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ribonucleótidos/farmacología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR
18.
Exp Biol Med (Maywood) ; 230(9): 606-11, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16179728

RESUMEN

Skeletal muscle serves as the body's major glutamine repository, and releases glutamine at enhanced rates during diabetes, but whether all muscles are equally affected is unknown. System N(m) activity mediates most trans-sarcolemmal glutamine movement, and although two System N (SN) isoforms have been identified (SN1/sodium-coupled neutral amino acid transporter or System N and A transporters [SNAT]-3; and SN2/SNAT5), their expression in skeletal muscle remains controversial. Here, the impact of Type I diabetes on glutamine uptake and System N transporter expression were examined in fast- and slow-twitch skeletal muscle from spontaneously diabetic (BB/Wor-DP) rats. Net glutamine uptake in fast-twitch fibers was decreased 75%-95%, but enhanced more than 2-fold in slow-twitch muscle from diabetic animals relative to nondiabetic controls. Both SNAT3 and SNAT5 mRNA were expressed in both muscle fiber types and their abundance was unaffected by diabetes. This represents the first report of differential fiber-specific effects of diabetes on skeletal muscle glutamine transport and the co-expression of distinct System N transporters in skeletal muscle.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Glutamina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Secuencia de Bases , ADN Complementario , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Ratas , Homología de Secuencia de Ácido Nucleico
19.
Am J Physiol Endocrinol Metab ; 289(6): E986-92, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16030062

RESUMEN

We hypothesized that AMP-activated protein kinase-related kinase 5 (ARK5)/novel kinase family 1 (NUAK1), an AMP-activated protein kinase (AMPK)-related kinase that has been found to be stimulated by protein kinase B (Akt), would be expressed in rat skeletal muscle and activated by electrically elicited contractions, 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR), or insulin. We verified expression of ARK5 in muscle through RT-PCR and Western blot. Cross-reactivity of ARK5 immunoprecipitates with antibodies against phospho-AMPK was increased by approximately 30% by muscle contractions and approximately 60% by incubation of muscle with AICAR. AMPK was not detected in the ARK5 immunoprecipitates. Despite the apparent increase in phosphorylation of ARK5 at a site essential to its activation, neither contractions nor AICAR increased ARK5 activity. For muscles from animals injected with saline or insulin, we probed nonimmunoprecipitated samples in sequence for phosphotyrosine (P-Tyr), ARK5, and phosphorylated substrates of Akt (P-AS) and found that the ARK5 band could be precisely superimposed on phosphoprotein bands from the P-Tyr and P-AS blots. In the band corresponding to ARK5, insulin increased P-Tyr content by approximately 45% and cross-reactivity with the antibody against P-AS by approximately threefold. We also detected ARK5 in phosphotyrosine immunoprecipitates. Our data suggest that increased phosphorylation of ARK5 by muscle contractions or exposure to AICAR is insufficient to activate ARK5 in skeletal muscle, suggesting that some other modification (e.g., phosphorylation on tyrosine or by Akt) may be necessary to its activity in muscle.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Hipoglucemiantes/farmacología , Insulina/farmacología , Contracción Muscular/fisiología , Músculo Esquelético/enzimología , Proteínas Quinasas/metabolismo , Proteínas Represoras/metabolismo , Ribonucleótidos/farmacología , Secuencia de Aminoácidos , Aminoimidazol Carboxamida/farmacología , Animales , Sitios de Unión , Western Blotting , Activación Enzimática/efectos de los fármacos , Expresión Génica , Técnicas de Inmunoadsorción , Masculino , Datos de Secuencia Molecular , Fosforilación , Proteínas Quinasas/química , Proteínas Quinasas/genética , ARN Mensajero/análisis , Ratas , Ratas Wistar , Proteínas Represoras/química , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Am J Physiol Endocrinol Metab ; 288(2): E347-52, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15494613

RESUMEN

The purpose of this study was to investigate the potential role of creatine in GLUT4 gene expression in rat skeletal muscle. Female Wistar rats were fed normal rat chow (controls) or chow containing 2% creatine monohydrate ad libitum for 3 wk. GLUT4 protein levels of creatine-fed rats were significantly increased in extensor digitorum longus (EDL), triceps, and epitrochlearis muscles compared with muscles from controls (P < 0.05), and triceps GLUT4 mRNA levels were approximately 100% greater in triceps muscles from creatine-fed rats than in muscles from controls (P < 0.05). In epitrochlearis muscles from creatine-fed animals, glycogen content was approximately 40% greater (P < 0.05), and insulin-stimulated glucose transport rates were higher (P < 0.05) than in epitrochlearis muscles from controls. Despite no changes in [ATP], [creatine], [phosphocreatine], or [AMP], creatine feeding increased AMP-activated protein kinase (AMPK) phosphorylation by 50% in rat EDL muscle (P < 0.05). Creatinine content of EDL muscle was almost twofold higher for creatine-fed animals than for controls (P < 0.05). Creatine feeding increased protein levels of myocyte enhancer factor 2 (MEF2) isoforms MEF2A ( approximately 70%, P < 0.05), MEF2C ( approximately 60%, P < 0.05), and MEF2D ( approximately 90%, P < 0.05), which are transcription factors that regulate GLUT4 expression, in creatine-fed rat EDL muscle nuclear extracts. Electrophoretic mobility shift assay showed that DNA binding activity of MEF2 was increased by approximately 40% (P < 0.05) in creatine-fed rat EDL compared with controls. Our data suggest that creatine feeding enhances the nuclear content and DNA binding activity of MEF2 isoforms, which is concomitant with an increase in GLUT4 gene expression.


Asunto(s)
Administración Oral , Creatina/administración & dosificación , Proteínas de Unión al ADN/metabolismo , Glucosa/metabolismo , Proteínas de Transporte de Monosacáridos/biosíntesis , Complejos Multienzimáticos/metabolismo , Proteínas Musculares/biosíntesis , Músculo Esquelético/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Transportador de Glucosa de Tipo 4 , Factores de Transcripción MEF2 , Músculo Esquelético/efectos de los fármacos , Factores Reguladores Miogénicos , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA