Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 854
Filter
1.
Biochem Biophys Res Commun ; 540: 116-122, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33472133

ABSTRACT

Mitochondrial dysfunction is considered to be a major cause of sarcopenia, defined as age-related muscle fiber atrophy and muscle weakness, as reduced mitochondrial respiration and morphological changes such as ragged red fibers (RRFs) are observed in aging muscles. However, the role of mitochondrial dysfunction in sarcopenia is not fully elucidated. Although previous studies have suggested that aging has a fiber type-specific effect on mitochondrial function, little is known about mitochondrial changes in individual fiber types. Here, we used C57BL/6NCr female mice to identify fiber type-specific pathological changes, examine the significance of pathological changes in sarcopenia, and identify possible mechanisms behind mitochondrial changes in slow-twitch soleus muscle (SOL) and fast-twitch extensor digitorum longus muscle (EDL). We observed reduced type I fiber-specific mitochondrial respiratory enzyme activity, impaired respiration, and subsarcolemmal mitochondrial accumulation in aged SOL, which was different from RRFs. These pathological alterations were not directly associated with fiber atrophy. Additionally, we found increased oxidative stress markers in aged SOL, suggesting that oxidative stress is involved in the pathological and functional changes in mitochondria. Meanwhile, obvious mitochondrial changes were not seen in aged EDL. Thus, age-related mitochondrial dysfunction is specific to the fiber type and may correlate with the muscle quality rather than the muscle mass.


Subject(s)
Aging/metabolism , Aging/pathology , Cell Respiration , Mitochondria/metabolism , Mitochondria/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Animals , Female , Mice , Mitochondria/enzymology , Muscle Fibers, Skeletal/enzymology , Organ Size , Oxidative Phosphorylation , Oxidative Stress , Sarcolemma/enzymology , Sarcolemma/metabolism , Sarcolemma/pathology , Sarcopenia/enzymology , Sarcopenia/metabolism , Sarcopenia/pathology
2.
Cell Death Dis ; 10(5): 354, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31043586

ABSTRACT

Neuronal nitric oxide synthase (nNOS) plays a crucial role in the maintenance of correct skeletal muscle function due, at least in part, to S-nitrosylation of specific protein targets. Similarly, we recently provided evidence for a muscular phenotype in mice lacking the denitrosylase S-nitrosoglutathione reductase (GSNOR). Here, we demonstrate that nNOS and GSNOR are concomitantly expressed during differentiation of C2C12. They colocalizes at the sarcolemma and co-immunoprecipitate in cells and in myofibers. We also provide evidence that GSNOR expression decreases in mouse models of muscular dystrophies and of muscle atrophy and wasting, i.e., aging and amyotrophic lateral sclerosis, suggesting a more general regulatory role of GSNOR in skeletal muscle homeostasis.


Subject(s)
Aging/genetics , Alcohol Dehydrogenase/genetics , Homeostasis/genetics , Muscle Development/genetics , Muscular Dystrophies/genetics , Nitric Oxide Synthase Type I/genetics , Aging/metabolism , Alcohol Dehydrogenase/antagonists & inhibitors , Alcohol Dehydrogenase/deficiency , Animals , Cell Differentiation , Cell Line, Transformed , Disease Models, Animal , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Gene Expression Regulation , Humans , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Myoblasts/cytology , Myoblasts/enzymology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sarcolemma/enzymology , Signal Transduction , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
3.
PLoS One ; 14(2): e0212219, 2019.
Article in English | MEDLINE | ID: mdl-30735546

ABSTRACT

Translocation of the glucose transporter GLUT4 to the sarcolemma accounts for glucose uptake in skeletal muscle following insulin administration. The protein kinase Akt2 and the small GTPase Rac1 have been implicated as essential regulators of insulin-stimulated GLUT4 translocation. Several lines of evidence suggest that Rac1 is modulated downstream of Akt2, and indeed the guanine nucleotide exchange factor FLJ00068 has been identified as an activator of Rac1. On the other hand, the mechanisms whereby Akt2 and Rac1 are regulated in parallel downstream of phosphoinositide 3-kinase are also proposed. Herein, we aimed to provide additional evidence that support a critical role for Akt2 in insulin regulation of Rac1 in mouse skeletal muscle. Knockdown of Akt2 by RNA interference abolished Rac1 activation following intravenous administration of insulin or ectopic expression of a constitutively activated phosphoinositide 3-kinase mutant. The activation of another small GTPase RalA and GLUT4 translocation to the sarcolemma following insulin administration or ectopic expression of a constitutively activated form of phosphoinositide 3-kinase, but not Rac1, were also diminished by downregulation of Akt2 expression. Collectively, these results strongly support the notion that Rac1 acts downstream of Akt2 leading to the activation of RalA and GLUT4 translocation to the sarcolemma in skeletal muscle.


Subject(s)
Glucose/metabolism , Insulin/pharmacology , Muscle, Skeletal/metabolism , Neuropeptides/metabolism , Proto-Oncogene Proteins c-akt/biosynthesis , Sarcolemma/enzymology , rac1 GTP-Binding Protein/metabolism , Animals , Down-Regulation/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Male , Mice , Mutation , Neuropeptides/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Sarcolemma/genetics , rac1 GTP-Binding Protein/genetics , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism
4.
Cardiovasc Res ; 115(3): 546-555, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30165515

ABSTRACT

AIMS: Cyclic adenosine monophosphate (cAMP) regulates cardiac excitation-contraction coupling by acting in microdomains associated with sarcolemmal ion channels. However, local real time cAMP dynamics in such microdomains has not been visualized before. We sought to directly monitor cAMP in a microdomain formed around sodium-potassium ATPase (NKA) in healthy and failing cardiomyocytes and to better understand alterations of cAMP compartmentation in heart failure. METHODS AND RESULTS: A novel Förster resonance energy transfer (FRET)-based biosensor termed phospholemman (PLM)-Epac1 was developed by fusing a highly sensitive cAMP sensor Epac1-camps to the C-terminus of PLM. Live cell imaging in PLM-Epac1 and Epac1-camps expressing adult rat ventricular myocytes revealed extensive regulation of NKA/PLM microdomain-associated cAMP levels by ß2-adrenoceptors (ß2-ARs). Local cAMP pools stimulated by these receptors were tightly controlled by phosphodiesterase (PDE) type 3. In chronic heart failure following myocardial infarction, dramatic reduction of the microdomain-specific ß2-AR/cAMP signals and ß2-AR dependent PLM phosphorylation was accompanied by a pronounced loss of local PDE3 and an increase in PDE2 effects. CONCLUSIONS: NKA/PLM complex forms a distinct cAMP microdomain which is directly regulated by ß2-ARs and is under predominant control by PDE3. In heart failure, local changes in PDE repertoire result in blunted ß2-AR signalling to cAMP in the vicinity of PLM.


Subject(s)
Cyclic AMP/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/enzymology , Phosphoproteins/metabolism , Receptors, Adrenergic, beta-2/metabolism , Sarcolemma/enzymology , Second Messenger Systems , Sodium-Potassium-Exchanging ATPase/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Biosensing Techniques , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Disease Models, Animal , Guanine Nucleotide Exchange Factors/metabolism , Heart Failure/enzymology , Heart Failure/pathology , Heart Failure/physiopathology , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Protein Interaction Domains and Motifs , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-2/drug effects , Sarcolemma/drug effects , Sarcolemma/pathology , Second Messenger Systems/drug effects , Time Factors
5.
Neuromuscul Disord ; 28(11): 914-926, 2018 11.
Article in English | MEDLINE | ID: mdl-30352768

ABSTRACT

Neuronal nitric oxide synthase (nNOS) is involved in nitric oxide (NO) production and suggested to play a crucial role in blood flow regulation of skeletal muscle. During activation of the muscle, NO helps attenuate the sympathetic vasoconstriction to accommodate increased metabolic demands, a phenomenon known as functional sympatholysis. In inherited myopathies such as the dystrophinopathies Duchenne and Becker muscle dystrophies (DMD and BMD), nNOS is lost from the sarcolemma. The loss of nNOS may cause functional ischemia contributing to skeletal and cardiac muscle cell injury. Effects of NO is augmented by inhibiting degradation of the second messenger cyclic guanosine monophosphate (cGMP) using sildenafil and tadalafil, both of which inhibit the enzyme phosphodiesterase 5 (PDE5). In animal models of DMD, PDE5-inhibitors prevent functional ischemia, reduce post-exercise skeletal muscle pathology and fatigue, show amelioration of cardiac muscle cell damage and increase cardiac performance. However, effect on clinical outcomes in DMD and BMD patients have been disappointing with minor effects on upper limb performance and none on ambulation. This review aims to summarize the current knowledge of nNOS function related to functional sympatholysis in skeletal muscle and studies on PDE5-inhibitor treatment in nNOS-deficient animal models and patients.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Nitric Oxide Synthase Type I/metabolism , Dystrophin/metabolism , Humans , Muscle, Skeletal/blood supply , Sarcolemma/enzymology , Signal Transduction/physiology
6.
J Pathol ; 246(4): 433-446, 2018 12.
Article in English | MEDLINE | ID: mdl-30066461

ABSTRACT

Skeletal muscle atrophy following unloading or immobilization represents a major invalidating event in bedridden patients. Among mechanisms involved in atrophy development, a controversial role is played by neuronal NOS (nNOS; NOS1), whose dysregulation at the protein level and/or subcellular distribution also characterizes other neuromuscular disorders. This study aimed to investigate unloading-induced changes in nNOS before any evidence of myofiber atrophy, using vastus lateralis biopsies obtained from young healthy subjects after a short bed-rest and rat soleus muscles after exposure to short unloading periods. Our results showed that (1) changes in nNOS subcellular distribution using NADPH-diaphorase histochemistry to detect enzyme activity were observed earlier than using immunofluorescence to visualize the protein; (2) loss of active nNOS from the physiological subsarcolemmal localization occurred before myofiber atrophy, i.e. in 8-day bed-rest biopsies and in 6 h-unloaded rat soleus, and was accompanied by increased nNOS activity in the sarcoplasm; (3) nNOS (Nos1) transcript and protein levels decreased significantly in the rat soleus after 6 h and 1 day unloading, respectively, to return to ambulatory levels after 4 and 7 days of unloading, respectively; (4) unloading-induced nNOS redistribution appeared dependent on mitochondrial-derived oxidant species, indirectly measured by tropomyosin disulfide bonds which had increased significantly in the rat soleus already after a 6 h-unloading bout; (5) activity of displaced nNOS molecules is required for translocation of the FoxO3 transcription factor to myofiber nuclei. FoxO3 nuclear localization in rat soleus increased after 6 h unloading (about four-fold the ambulatory level), whereas it did not when nNOS expression and activity were inhibited in vivo before and during 6 h unloading. In conclusion, this study demonstrates that the redistribution of active nNOS molecules from sarcolemma to sarcoplasm not only is ahead of the atrophy of unloaded myofibers, and is induced by increased production of mitochondrial superoxide anion, but also drives FoxO3 activation to initiate muscle atrophy. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Muscular Atrophy/enzymology , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress , Quadriceps Muscle/enzymology , Sarcolemma/enzymology , Animals , Bed Rest , Disease Models, Animal , Down-Regulation , Female , Forkhead Box Protein O3/metabolism , Healthy Volunteers , Hindlimb Suspension , Humans , Male , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , NADP/metabolism , Nitric Oxide Synthase Type I/genetics , Protein Transport , Quadriceps Muscle/pathology , Quadriceps Muscle/physiopathology , Rats, Wistar , Sarcolemma/pathology , Superoxides/metabolism , Time Factors
7.
Am J Physiol Cell Physiol ; 313(6): C604-C611, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28971834

ABSTRACT

Mechanistic target of rapamycin (mTOR) resides as two complexes within skeletal muscle. mTOR complex 1 [mTORC1-regulatory associated protein of mTOR (Raptor) positive] regulates skeletal muscle growth, whereas mTORC2 [rapamycin-insensitive companion of mTOR (Rictor) positive] regulates insulin sensitivity. To examine the regulation of these complexes in human skeletal muscle, we utilized immunohistochemical analysis to study the localization of mTOR complexes before and following protein-carbohydrate feeding (FED) and resistance exercise plus protein-carbohydrate feeding (EXFED) in a unilateral exercise model. In basal samples, mTOR and the lysosomal marker lysosomal associated membrane protein 2 (LAMP2) were highly colocalized and remained so throughout. In the FED and EXFED states, mTOR/LAMP2 complexes were redistributed to the cell periphery [wheat germ agglutinin (WGA)-positive staining] (time effect; P = 0.025), with 39% (FED) and 26% (EXFED) increases in mTOR/WGA association observed 1 h post-feeding/exercise. mTOR/WGA colocalization continued to increase in EXFED at 3 h (48% above baseline) whereas colocalization decreased in FED (21% above baseline). A significant effect of condition (P = 0.05) was noted suggesting mTOR/WGA colocalization was greater during EXFED. This pattern was replicated in Raptor/WGA association, where a significant difference between EXFED and FED was noted at 3 h post-exercise/feeding (P = 0.014). Rictor/WGA colocalization remained unaltered throughout the trial. Alterations in mTORC1 cellular location coincided with elevated S6K1 kinase activity, which rose to a greater extent in EXFED compared with FED at 1 h post-exercise/feeding (P < 0.001), and only remained elevated in EXFED at the 3 h time point (P = 0.037). Collectively these data suggest that mTORC1 redistribution within the cell is a fundamental response to resistance exercise and feeding, whereas mTORC2 is predominantly situated at the sarcolemma and does not alter localization.


Subject(s)
Eating , Energy Metabolism , Exercise , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Quadriceps Muscle/enzymology , Adult , Dietary Carbohydrates/administration & dosage , Dietary Proteins/administration & dosage , Humans , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomes/enzymology , Male , Muscle Contraction , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Resistance Training , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sarcolemma/enzymology , Time Factors , Young Adult
8.
BMC Complement Altern Med ; 16: 209, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27405506

ABSTRACT

BACKGROUND: Rhodiola species have been used for asthenia, depression, fatigue, poor work performance and cardiovascular diseases, all of which may be associated with insulin resistance. To disclose the underlying mechanisms of action, the effect of Rhodiola crenulata root (RCR) on insulin resistance was investigated. METHODS: Male Sprague-Dawley rats were treated with liquid fructose in their drinking water over 18 weeks. The extract of RCR was co-administered (once daily by oral gavage) during the last 5 weeks. The indexes of lipid and glucose homeostasis were determined enzymatically and/or by ELISA. Gene expression was analyzed by Real-time PCR, Western blot and/or confocal immunofluorescence. RESULTS: RCR extract (50 mg/kg) suppressed fructose-induced hyperinsulinemia and the increases in the homeostasis model assessment of insulin resistance index and the adipose tissue insulin resistance index in rats. Additionally, this treatment had a trend to restore the ratios of glucose to insulin and non-esterified fatty acids (NEFA) to insulin. Mechanistically, RCR suppressed fructose-induced acceleration of the clearance of plasma NEFA during oral glucose tolerance test (OGTT), and decreased triglyceride content and Oil Red O staining area in the gastrocnemius. Furthermore, RCR restored fructose-induced sarcolemmal overexpression and intracellular less distribution of fatty acid translocase/CD36 that contributes to etiology of insulin resistance by facilitating fatty acid uptake. CONCLUSION: These results suggest that RCR ameliorates insulin resistance in fructose-fed rats by modulating sarcolemmal and intracellular CD36 redistribution in the skeletal muscle. Our findings may provide a better understanding of the traditional use of Rhodila species.


Subject(s)
CD36 Antigens/metabolism , Insulin Resistance , Plant Extracts/pharmacology , Plant Roots/chemistry , Rhodiola/chemistry , Animals , Fructose/administration & dosage , Lipid Metabolism , Male , Rats , Rats, Sprague-Dawley , Sarcolemma/enzymology
9.
J Biol Chem ; 291(19): 9920-8, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26966179

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by mutations in the gene encoding dystrophin. Loss of dystrophin protein compromises the stability of the sarcolemma membrane surrounding each muscle cell fiber, leading to membrane ruptures and leakiness that induces myofiber necrosis, a subsequent inflammatory response, and progressive tissue fibrosis with loss of functional capacity. Cathepsin S (Ctss) is a cysteine protease that is actively secreted in areas of tissue injury and ongoing inflammation, where it participates in extracellular matrix remodeling and healing. Here we show significant induction of Ctss expression and proteolytic activity following acute muscle injury or in muscle from mdx mice, a model of DMD. To examine the functional ramifications associated with greater Ctss expression, the Ctss gene was deleted in the mdx genetic background, resulting in protection from muscular dystrophy pathogenesis that included reduced myofiber turnover and histopathology, reduced fibrosis, and improved running capacity. Mechanistically, deletion of the Ctss gene in the mdx background significantly increased myofiber sarcolemmal membrane stability with greater expression and membrane localization of utrophin, integrins, and ß-dystroglycan, which anchor the membrane to the basal lamina and underlying cytoskeletal proteins. Consistent with these results, skeletal muscle-specific transgenic mice overexpressing Ctss showed increased myofiber necrosis, muscle histopathology, and a functional deficit reminiscent of muscular dystrophy. Hence, Ctss induction during muscular dystrophy is a pathologic event that partially underlies disease pathogenesis, and its inhibition might serve as a new therapeutic strategy in DMD.


Subject(s)
Cathepsins/biosynthesis , Gene Expression Regulation, Developmental , Muscle Fibers, Skeletal/enzymology , Muscular Dystrophy, Animal/enzymology , Muscular Dystrophy, Duchenne/enzymology , Animals , Cytoskeleton/enzymology , Cytoskeleton/genetics , Cytoskeleton/pathology , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Necrosis , Proteolysis , Sarcolemma/enzymology , Sarcolemma/genetics , Sarcolemma/pathology
10.
Neuromuscul Disord ; 25(12): 964-76, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26483274

ABSTRACT

The neuronal nitric-oxide synthase (nNOS) splice variant nNOSµ is essential for skeletal muscle function. Its localization is dependent on dystrophin, which stabilizes the dystrophin glycoprotein complex (DGC) at the sarcolemma of skeletal muscle fibers. In Duchenne muscular dystrophy (DMD) dystrophin is absent and sarcolemmal nNOS is lost. This leads to functional ischemia due to a decrease in contraction-induced vasodilation. In cardiomyocytes, nNOSµ is believed to be the predominant NOS isoform. However, the association of nNOS with the DGC in the heart is unclear. Here, we report nNOS localization at the intercalated discs (IDs) of cardiomyocytes, where utrophin is highly expressed. In mdx, mdx:utr, nNOSµ knock-out (KO), and mdx:nNOSµ KO mice, we observed a gradual reduction of nNOS at IDs and disrupted ID morphology, compared to wild-type. In mdx:nNOSµ KO mice, but not in mdx or nNOSµ KO mice, we also observed an early development of cardiac fibrosis. These findings suggest that nNOS localization in the heart may not depend exclusively on the presence of dystrophin. Additionally, the ß1 subunit of soluble guanylyl cyclase (sGC), responsible for the production of cGMP through nitric oxide (NO) signaling, was also detected at the IDs. Together, our results suggest a new role of nNOS at the IDs for the cGMP-dependent NO pathway and the maintenance of ID morphology.


Subject(s)
Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Nitric Oxide Synthase Type I/metabolism , Utrophin/metabolism , Animals , Guanylate Cyclase/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Nitric Oxide Synthase Type I/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sarcolemma/enzymology , Soluble Guanylyl Cyclase , Utrophin/genetics
11.
Am J Physiol Endocrinol Metab ; 308(8): E613-20, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25670828

ABSTRACT

Development of acute insulin resistance represents a negative factor after surgery, but the underlying mechanisms are not fully understood. We investigated the postoperative changes in insulin sensitivity, mitochondrial function, enzyme activities, and release of reactive oxygen species (ROS) in skeletal muscle and liver in pigs on the 2nd postoperative day after major abdominal surgery. Peripheral and hepatic insulin sensitivity were assessed by D-[6,6-²H2]glucose infusion and hyperinsulinemic euglycemic step clamping. Surgical trauma elicited a decline in peripheral insulin sensitivity (∼34%, P<0.01), whereas hepatic insulin sensitivity remained unchanged. Intramyofibrillar (IFM) and subsarcolemma mitochondria (SSM) isolated from skeletal muscle showed a postoperative decline in ADP-stimulated respiration (V(ADP)) for pyruvate (∼61%, P<0.05, and ∼40%, P<0.001, respectively), whereas V(ADP) for glutamate and palmitoyl-L-carnitine (PC) was unchanged. Mitochondrial leak respiration with PC was increased in SSM (1.9-fold, P<0.05) and IFM (2.5-fold, P<0.05), indicating FFA-induced uncoupling. The activity of the pyruvate dehydrogenase complex (PDC) was reduced (∼32%, P<0.01) and positively correlated to the decline in peripheral insulin sensitivity (r=0.748, P<0.05). All other mitochondrial enzyme activities were unchanged. No changes in mitochondrial function in liver were observed. Mitochondrial H2O2 and O2·â» emission was measured spectrofluorometrically, and H2O2 was increased in SSM, IFM, and liver mitochondria (∼2.3-, ∼2.5-, and ∼2.3-fold, respectively, all P<0.05). We conclude that an impairment in skeletal muscle mitochondrial PDC activity and pyruvate oxidation capacity arises in the postoperative phase along with increased ROS emission, suggesting a link between mitochondrial function and development of acute postoperative insulin resistance.


Subject(s)
Insulin Resistance , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Oxidative Stress , Postoperative Complications/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Reactive Oxygen Species/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Crosses, Genetic , Glucose Clamp Technique , Liver/enzymology , Liver/metabolism , Mitochondria, Liver/enzymology , Mitochondria, Liver/metabolism , Mitochondria, Muscle/enzymology , Muscle, Skeletal/enzymology , Myofibrils/enzymology , Myofibrils/metabolism , Organ Specificity , Oxidative Phosphorylation , Oxygen Consumption , Postoperative Complications/blood , Postoperative Complications/enzymology , Pyruvic Acid/metabolism , Sarcolemma/enzymology , Sarcolemma/metabolism , Sus scrofa
12.
J Physiol ; 592(21): 4627-38, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25194047

ABSTRACT

The secondary loss of neuronal nitric oxide synthase (nNOS) that occurs in dystrophic muscle is the basis of numerous, complex and interacting features of the dystrophic pathology that affect not only muscle itself, but also influence the interaction of muscle with other tissues. Many mechanisms through which nNOS deficiency contributes to misregulation of muscle development, blood flow, fatigue, inflammation and fibrosis in dystrophic muscle have been identified, suggesting that normalization in NO production could greatly attenuate diverse aspects of the pathology of muscular dystrophy through multiple regulatory pathways. However, the relative importance of the loss of nNOS from the sarcolemma versus the importance of loss of total nNOS from dystrophic muscle remains unknown. Although most current evidence indicates that nNOS localization at the sarcolemma is not required to achieve NO-mediated reductions of pathology in muscular dystrophy, the question remains open concerning whether membrane localization would provide a more efficient rescue from features of the dystrophic phenotype.


Subject(s)
Muscular Dystrophy, Duchenne/enzymology , Muscular Dystrophy, Duchenne/physiopathology , Nitric Oxide Synthase Type I/deficiency , Animals , Dystrophin/deficiency , Dystrophin/genetics , Dystrophin/metabolism , Humans , Nitric Oxide/metabolism , Sarcolemma/enzymology
13.
Basic Res Cardiol ; 109(4): 414, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24816396

ABSTRACT

Hypoxic preconditioning (HPC) protects rat hearts against ischemia-reperfusion (IR) injury. However, the role of transient receptor potential vanilloid 1 (TRPV1) in HPC-mediated cardioprotection remains unknown. TRPV1 is activated by endovanilloid 12(S)-hydroxyeicosatetraenoic acid [12(S)-HETE], which is synthesized by arachidonate 12-lipoxygenase (ALOX12). Therefore, we examined whether HPC protects the myocardium against IR via the ALOX12/TRPV1 pathway. Compared to hearts of rats kept in room air, the hearts of rats kept in air with 10 % oxygen for 4 weeks had better post-ischemic recovery and less tissue damage when subjected to 30-min global ischemia and 4-h reflow in a Langendorff apparatus. Capsazepine, a specific TRPV1 blocker, administered 5 min before reperfusion markedly attenuated the effects of HPC, confirming that TRPV1 is a downstream effector in HPC-mediated cardioprotection. HPC resulted in the upregulation of ALOX12 and myocardial 12(S)-HETE, and prevented IR-induced 12(S)-HETE reduction. In addition, sarcolemmal ALOX12 expression in HPC hearts mainly co-localized with TRPV1 expression. Blockade of ALOX12 by cinnamyl-3,4-dihydroxy-α-cyanocinnamate or baicalein abrogated the effects of HPC, baicalein also decreased 12(S)-HETE expression. Mimicking HPC by given 12(S)-HETE or capsaicin to baicalien-treated hearts enhanced cardiac recovery during reperfusion. The cardiac protein kinase C (PKC) isoforms α, δ, ε, and ζ were preferentially expressed in the sarcolemmal membrane of HPC-treated hearts, indicating their high intrinsic activation state. Capsazepine or co-treatment with baicalein attenuated translocation of PKCα, PKCδ and PKCε, but not that of PKCζ. We conclude that HPC reduces heart susceptibly to IR via ALOX12/TRPV1/PKC pathway, as shown by increased 12(S)-HETE expression in HPC hearts.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Hypoxia/enzymology , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Signal Transduction , TRPV Cation Channels/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Animals , Caffeic Acids/pharmacology , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Flavanones/pharmacology , In Vitro Techniques , Isoenzymes , Lipoxygenase Inhibitors/pharmacology , Male , Myocardial Reperfusion Injury/enzymology , Protein Kinase C/metabolism , Protein Transport , Rats, Wistar , Sarcolemma/enzymology , Signal Transduction/drug effects , TRPV Cation Channels/antagonists & inhibitors , Time Factors
14.
Am J Physiol Regul Integr Comp Physiol ; 306(7): R470-82, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24477538

ABSTRACT

Reduced mechanical loading during bedrest, spaceflight, and casting, causes rapid morphological changes in skeletal muscle: fiber atrophy and reduction of slow-twitch fibers. An emerging signaling event in response to unloading is the translocation of neuronal nitric oxide synthase (nNOSµ) from the sarcolemma to the cytosol. We used EUK-134, a cell-permeable mimetic of superoxide dismutase and catalase, to test the role of redox signaling in nNOSµ translocation and muscle fiber atrophy as a result of short-term (54 h) hindlimb unloading. Fischer-344 rats were divided into ambulatory control, hindlimb-unloaded (HU), and hindlimb-unloaded + EUK-134 (HU-EUK) groups. EUK-134 mitigated the unloading-induced phenotype, including muscle fiber atrophy and muscle fiber-type shift from slow to fast. nNOSµ immunolocalization at the sarcolemma of the soleus was reduced with HU, while nNOSµ protein content in the cytosol increased with unloading. Translocation of nNOS from the sarcolemma to cytosol was virtually abolished by EUK-134. EUK-134 also mitigated dephosphorylation at Thr-32 of FoxO3a during HU. Hindlimb unloading elevated oxidative stress (4-hydroxynonenal) and increased sarcolemmal localization of Nox2 subunits gp91phox (Nox2) and p47phox, effects normalized by EUK-134. Thus, our findings are consistent with the hypothesis that oxidative stress triggers nNOSµ translocation from the sarcolemma and FoxO3a dephosphorylation as an early event during mechanical unloading. Thus, redox signaling may serve as a biological switch for nNOS to initiate morphological changes in skeletal muscle fibers.


Subject(s)
Antioxidants/pharmacology , Hindlimb Suspension , Muscle Fibers, Skeletal/drug effects , Muscular Atrophy/prevention & control , Nitric Oxide Synthase Type I/metabolism , Organometallic Compounds/pharmacology , Salicylates/pharmacology , Aldehydes/metabolism , Animals , Cytosol/drug effects , Cytosol/enzymology , Disease Models, Animal , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Membrane Glycoproteins/metabolism , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/enzymology , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/enzymology , Muscle Fibers, Slow-Twitch/pathology , Muscular Atrophy/enzymology , Muscular Atrophy/pathology , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Phenotype , Phosphorylation , Protein Transport , Rats , Rats, Inbred F344 , Sarcolemma/drug effects , Sarcolemma/enzymology , Signal Transduction/drug effects , Time Factors
15.
J Mol Cell Cardiol ; 68: 47-55, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412535

ABSTRACT

L-type Ca channels (LTCC), which play a key role in cardiac excitation-contraction coupling, are located predominantly at the transverse (t-) tubules in ventricular myocytes. Caveolae and the protein caveolin-3 (Cav-3) are also present at the t-tubules and have been implicated in localizing a number of signaling molecules, including protein kinase A (PKA) and ß2-adrenoceptors. The present study investigated whether disruption of Cav-3 binding to its endogenous binding partners influenced LTCC activity. Ventricular myocytes were isolated from male Wistar rats and LTCC current (ICa) recorded using the whole-cell patch-clamp technique. Incubation of myocytes with a membrane-permeable peptide representing the scaffolding domain of Cav-3 (C3SD) reduced basal ICa amplitude in intact, but not detubulated, myocytes, and attenuated the stimulatory effects of the ß2-adrenergic agonist zinterol on ICa. The PKA inhibitor H-89 also reduced basal ICa; however, the inhibitory effects of C3SD and H-89 on basal ICa amplitude were not summative. Under control conditions, myocytes stained with antibody against phosphorylated LTCC (pLTCC) displayed a striated pattern, presumably reflecting localization at the t-tubules. Both C3SD and H-89 reduced pLTCC staining at the z-lines but did not affect staining of total LTCC or Cav-3. These data are consistent with the idea that the effects of C3SD and H-89 share a common pathway, which involves PKA and is maximally inhibited by H-89, and suggest that Cav-3 plays an important role in mediating stimulation of ICa at the t-tubules via PKA-induced phosphorylation under basal conditions, and in response to ß2-adrenoceptor stimulation.


Subject(s)
Calcium Signaling , Caveolin 3/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Ventricles/cytology , Myocytes, Cardiac/enzymology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Calcium Channels, L-Type/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Ethanolamines/pharmacology , Isoquinolines/pharmacology , Male , Patch-Clamp Techniques , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Rats , Rats, Wistar , Sarcolemma/enzymology , Sulfonamides/pharmacology
16.
J Cell Sci ; 127(Pt 5): 1033-42, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24413164

ABSTRACT

Multiple cAMP phosphodiesterase (PDE) isoforms play divergent roles in cardiac homeostasis but the molecular basis for their non-redundant function remains poorly understood. Here, we report a novel role for the PDE4B isoform in ß-adrenergic (ßAR) signaling in the heart. Genetic ablation of PDE4B disrupted ßAR-induced cAMP transients, as measured by FRET sensors, at the sarcolemma but not in the bulk cytosol of cardiomyocytes. This effect was further restricted to a subsarcolemmal compartment because PDE4B regulates ß1AR-, but not ß2AR- or PGE2-induced responses. The spatially restricted function of PDE4B was confirmed by its selective effects on PKA-mediated phosphorylation patterns. PDE4B limited the PKA-mediated phosphorylation of key players in excitation-contraction coupling that reside in the sarcolemmal compartment, including L-type Ca(2+) channels and ryanodine receptors, but not phosphorylation of distal cytosolic proteins. ß1AR- but not ß2AR-ligation induced PKA-dependent activation of PDE4B and interruption of this negative feedback with PKA inhibitors increased sarcolemmal cAMP. Thus, PDE4B mediates a crucial PKA-dependent feedback that controls ß1AR-dependent cAMP signals in a restricted subsarcolemmal domain. Disruption of this feedback augments local cAMP/PKA signals, leading to an increased intracellular Ca(2+) level and contraction rate.


Subject(s)
Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Myocytes, Cardiac/enzymology , Receptors, Adrenergic, beta-1/metabolism , Sarcolemma/enzymology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Animals , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Feedback, Physiological , Imidazoles/pharmacology , Myocardial Contraction , Phosphorylation , Protein Processing, Post-Translational , Rats , Receptors, Adrenergic, beta-2/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Second Messenger Systems
17.
Antioxid Redox Signal ; 20(16): 2479-96, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24093939

ABSTRACT

AIMS: Redox and growth-factor imbalance fosters muscle disuse atrophy. Since the endoplasmic-reticulum chaperone Grp94 is required for folding insulin-like growth factors (IGFs) and for antioxidant cytoprotection, we investigated its involvement in muscle mass loss due to inactivity. RESULTS: Rat soleus muscles were transfected in vivo and analyzed after 7 days of hindlimb unloading, an experimental model of muscle disuse atrophy, or standard caging. Increased muscle protein carbonylation and decreased Grp94 protein levels (p<0.05) characterized atrophic unloaded solei. Recombinant Grp94 expression significantly reduced atrophy of transfected myofibers, compared with untransfected and empty-vector transfected ones (p<0.01), and decreased the percentage of carbonylated myofibers (p=0.001). Conversely, expression of two different N-terminal deleted Grp94 species did not attenuate myofiber atrophy. No change in myofiber trophism was detected in transfected ambulatory solei. The absence of effects on atrophic untransfected myofibers excluded a major role for IGFs folded by recombinant Grp94. Immunoprecipitation and confocal microscopy assays to investigate chaperone interaction with muscle atrophy regulators identified 160 kDa neuronal nitric oxide synthase (nNOS) as a new Grp94 partner. Unloading was demonstrated to untether nNOS from myofiber subsarcolemma; here, we show that such nNOS localization, revealed by means of NADPH-diaphorase histochemistry, appeared preserved in unloaded myofibers expressing recombinant Grp94, compared to those transfected with the empty vector or deleted Grp94 cDNA (p<0.02). INNOVATION: Grp94 interacts with nNOS and prevents its untethering from sarcolemma in unloaded myofibers. CONCLUSION: Maintenance of Grp94 expression is sufficient to counter unloading atrophy and oxidative stress by mechanistically stabilizing nNOS-multiprotein complex at the myofiber sarcolemma.


Subject(s)
Membrane Glycoproteins/metabolism , Muscular Disorders, Atrophic/metabolism , Muscular Disorders, Atrophic/pathology , Nitric Oxide Synthase Type I/metabolism , Sarcolemma/enzymology , Animals , Enzyme Stability , Female , Muscular Disorders, Atrophic/enzymology , Rats , Rats, Wistar , Sarcolemma/metabolism
18.
Mol Cell Biochem ; 388(1-2): 241-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24347175

ABSTRACT

Previous studies showed that adverse effect of ionizing radiation on the cardiovascular system is beside other factors mostly mediated by reactive oxygen and nitrogen species, which deplete antioxidant stores. One of the structures highly sensitive to radicals is the Na,K-ATPase the main system responsible for extrusion of superfluous Na(+) out of the cell which utilizes the energy derived from ATP. The aim of present study was the investigation of functional properties of cardiac Na,K-ATPase in 20-week-old male rats 6 weeks after γ-irradiation by a dose 25 Gy (IR). Irradiation induced decrease of systolic blood pressure from 133 in controls to 85 mmHg in IR group together with hypertrophy of right ventricle (RV) and hypotrophy of left ventricle (LV). When activating the cardiac Na,K-ATPase with substrate, its activity was lower in IR in the whole concentration range of ATP. Evaluation of kinetic parameters revealed a decrease of the maximum velocity (V max) by 40 % with no changes in the value of Michaelis-Menten constant (K m). During activation with Na(+), we observed a decrease of the enzyme activity in hearts from IR at all tested Na(+) concentrations. The value of V max decreased by 38 %, and the concentration of Na(+) that gives half maximal reaction velocity (K Na) increased by 62 %. This impairment in the affinity of the Na(+)-binding site together with decreased number of active Na,K-ATPase molecules, as indicated by lowered V max values, are probably responsible for the deteriorated efflux of the excessive Na(+) from the intracellular space in hearts of irradiated rats.


Subject(s)
Gamma Rays/adverse effects , Heart/radiation effects , Sarcolemma/radiation effects , Sodium-Potassium-Exchanging ATPase/radiation effects , Animals , Blood Pressure/radiation effects , Hypertension/physiopathology , Hypertrophy, Left Ventricular , Hypertrophy, Right Ventricular , Male , Myocardium/enzymology , Neoplasms/radiotherapy , Rats , Rats, Wistar , Reactive Oxygen Species/adverse effects , Sarcolemma/enzymology
20.
Horm Metab Res ; 45(12): 874-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23918688

ABSTRACT

The purpose of the present study was to assess the impact of previous period of caloric restriction on energy balance and skeletal muscle mitochondrial energetics in response to high-fat (HF) diet. To this end, 1 group of rats was subjected to 2 weeks of caloric restriction with nonpurified diet and then fed HF diet (430 kJ metabolizable energy/day) for 1 week, while the second group was fed ad libitum with nonpurified diet for 2 weeks and then fed HF diet (430 kJ metabolizable energy/day) for 1 week. Body composition, energy balance, and glucose homeostasis were measured. Mitochondrial mass, oxidative capacity and efficiency, parameters of oxidative stress, and antioxidant defense were evaluated in subsarcolemmal and intermyofibrillar mitochondria from skeletal muscle. Body energy and lipid content, plasma insulin, and metabolic efficiency were significantly higher, while energy expenditure significantly decreased, in food-restricted rats fed HF diet compared to controls. Mitochondrial efficiency and oxidative damage in skeletal muscle were significantly increased, while antioxidant defence was significantly lower in food-restricted rats fed HF diet, compared with controls. Finally, food-restricted rats fed HF diet exhibited significant reduction in subsarcolemmal mitochondrial mass. In conclusion, caloric restriction elicits higher mitochondrial efficiency and predisposes skeletal muscle to high fat-induced oxidative damage, which in turn could lead to impaired glucose homeostasis in food-restricted rats fed HF diet.


Subject(s)
Caloric Restriction , Diet, High-Fat , Feeding Behavior , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology , Oxidative Stress , Aconitate Hydratase/metabolism , Animals , Blood Glucose/metabolism , Body Composition , Citrate (si)-Synthase/metabolism , Energy Metabolism , Insulin/blood , Kinetics , Lipid Peroxidation , Male , Mitochondria, Muscle/enzymology , Muscle, Skeletal/enzymology , Protons , Rats , Rats, Sprague-Dawley , Sarcolemma/enzymology , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...