Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Comput Biol Med ; 153: 106515, 2023 02.
Article in English | MEDLINE | ID: mdl-36610217

ABSTRACT

Transgelin-2 (TG2) is a novel promising therapeutic target for the treatment of asthma as it plays an important role in relaxing airway smooth muscles and reducing pulmonary resistance in asthma. The compound TSG12 is the only reported TG2 agonist with in vivo anti-asthma activity. However, the dynamic behavior and ligand binding sites of TG2 and its binding mechanism with TSG12 remain unclear. In this study, we performed 12.6 µs molecular dynamics (MD) simulations for apo-TG2 and TG2-TSG12 complex, respectively. The results suggested that the apo-TG2 has 4 most populated conformations, and that its binding of the agonist could expand the conformation distribution space of the protein. The simulations revealed 3 potential binding sites in 3 most populated conformations, one of which is induced by the agonist binding. Free energy decomposition uncovered 8 important residues with contributions stronger than -1 kcal/mol. Computational alanine scanning for the important residues by 100 ns conventional MD simulation for each mutated TG2-TSG12 complexes demonstrated that E27, R49 and F52 are essential residues for the agonist binding. These results should be helpful to understand the dynamic behavior of TG2 and its binding mechanism with the agonist TSG12, which could provide some structural insights into the novel mechanism for anti-asthma drug development.


Subject(s)
Anti-Asthmatic Agents , Molecular Dynamics Simulation , Anti-Asthmatic Agents/pharmacology , Muscle Proteins/agonists , Muscle Proteins/metabolism , Binding Sites , Drug Discovery , Protein Binding , Molecular Docking Simulation
2.
Nat Commun ; 13(1): 149, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013221

ABSTRACT

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Subject(s)
Cachexia/genetics , Endomyocardial Fibrosis/genetics , Heart Failure/genetics , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Transcription Factors/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , Aged , Aged, 80 and over , Animals , Cachexia/metabolism , Cachexia/physiopathology , Cachexia/prevention & control , Case-Control Studies , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/physiopathology , Endomyocardial Fibrosis/prevention & control , Female , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/prevention & control , Heart Function Tests , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/deficiency , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Muscular Atrophy/prevention & control , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency
3.
Trends Biochem Sci ; 44(10): 885-896, 2019 10.
Article in English | MEDLINE | ID: mdl-31256982

ABSTRACT

Transgelin-2 has been regarded as an actin-binding protein that induces actin gelation and regulates actin cytoskeleton. However, transgelin-2 has recently been shown to relax the myosin cytoskeleton of the airway smooth muscle cells by acting as a receptor for extracellular metallothionein-2. From a clinical perspective, these results support transgelin-2 as a promising therapeutic target for diseases such as cancer and asthma. The inhibition of transgelin-2 prevents actin gelation and thereby cancer cell proliferation, invasion, and metastasis. Conversely, the activation of transgelin-2 with specific agonists relaxes airway smooth muscles and reduces pulmonary resistance in asthma. Here, we review new studies on the biochemical properties of transgelin-2 and discuss their clinical implications for the treatment of immune, oncogenic, and respiratory disorders.


Subject(s)
Asthma/metabolism , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Neoplasms/metabolism , Actins/metabolism , Animals , Asthma/drug therapy , Asthma/pathology , Cell Proliferation/drug effects , Humans , Microfilament Proteins/agonists , Microfilament Proteins/antagonists & inhibitors , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology
4.
Sci Transl Med ; 10(427)2018 02 07.
Article in English | MEDLINE | ID: mdl-29437149

ABSTRACT

There is a clinical need for new bronchodilator drugs in asthma, because more than half of asthmatic patients do not receive adequate control with current available treatments. We report that inhibition of metallothionein-2 protein expression in lung tissues causes the increase of pulmonary resistance. Conversely, metallothionein-2 protein is more effective than ß2-agonists in reducing pulmonary resistance in rodent asthma models, alleviating tension in tracheal spirals, and relaxing airway smooth muscle cells (ASMCs). Metallothionein-2 relaxes ASMCs via transgelin-2 (TG2) and induces dephosphorylation of myosin phosphatase target subunit 1 (MYPT1). We identify TSG12 as a nontoxic, specific TG2-agonist that relaxes ASMCs and reduces asthmatic pulmonary resistance. In vivo, TSG12 reduces pulmonary resistance in both ovalbumin- and house dust mite-induced asthma in mice. TSG12 induces RhoA phosphorylation, thereby inactivating the RhoA-ROCK-MYPT1-MLC pathway and causing ASMCs relaxation. TSG12 is more effective than ß2-agonists in relaxing human ASMCs and pulmonary resistance with potential clinical advantages. These results suggest that TSG12 could be a promising therapeutic approach for treating asthma.


Subject(s)
Asthma/drug therapy , Asthma/metabolism , Lung/metabolism , Lung/pathology , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Animals , Disease Models, Animal , Mice , Mice, Knockout , Microfilament Proteins/agonists , Microfilament Proteins/genetics , Molecular Docking Simulation , Muscle Proteins/agonists , Muscle Proteins/genetics
5.
J Physiol Biochem ; 74(1): 35-45, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29198059

ABSTRACT

Caffeine has been shown to stimulate multiple major regulators of cell energetics including AMP-activated protein kinase (AMPK) and Ca2+/calmodulin-dependent protein kinase II (CaMKII). Additionally, caffeine induces peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and mitochondrial biogenesis. While caffeine enhances oxidative metabolism, experimental concentrations often exceed physiologically attainable concentrations through diet. This work measured the effects of low-level caffeine on cellular metabolism and gene expression in myotubes, as well as the dependence of caffeine's effects on the nuclear receptor peroxisome proliferator-activated receptor beta/delta (PPARß/δ). C2C12 myotubes were treated with various doses of caffeine for up to 24 h. Gene and protein expression were measured via qRT-PCR and Western blot, respectively. Cellular metabolism was determined via oxygen consumption and extracellular acidification rate. Caffeine significantly induced regulators of mitochondrial biogenesis and oxidative metabolism. Mitochondrial staining was suppressed in PPARß/δ-inhibited cells which was rescued by concurrent caffeine treatment. Caffeine-treated cells also displayed elevated peak oxidative metabolism which was partially abolished following PPARß/δ inhibition. Similar to past observations, glucose uptake and GLUT4 content were elevated in caffeine-treated cells, however, glycolytic metabolism was unaltered following caffeine treatment. Physiological levels of caffeine appear to enhance cell metabolism through mechanisms partially dependent on PPARß/δ.


Subject(s)
Caffeine/metabolism , Gene Expression Regulation , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , PPAR delta/agonists , PPAR-beta/agonists , Animals , Benzamides/pharmacology , Biological Assay , Cell Line , Coculture Techniques , Gene Expression Regulation/drug effects , Hydrogen-Ion Concentration , Lipid Metabolism/drug effects , Mice , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/enzymology , Mitochondrial Dynamics/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Organelle Biogenesis , Osmolar Concentration , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , PPAR delta/antagonists & inhibitors , PPAR delta/metabolism , PPAR-beta/antagonists & inhibitors , PPAR-beta/metabolism , Smegmamorpha , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Sulfones/pharmacology
6.
Peptides ; 99: 92-98, 2018 01.
Article in English | MEDLINE | ID: mdl-29183756

ABSTRACT

The results of our and other's studies showed that activation of galanin receptor 1 could mitigate insulin resistance via promoting glucose transporter 4 (GLUT4) expression and translocation in the skeletal muscle of rats. But no literature are available regarding the effect of galanin receptor 2 (GALR2) on insulin resistance in skeletal muscle of type 2 diabetes. Herein, in this study we intended to survey the effect of GALR2 and its signal mechanisms in the mice with high fat diet-induced obese. The mice were intraperitoneally injected with vehicle, GALR2 agonist M1145 and antagonist M871 respectively once a day for continuous 21 days. The skeletal muscles were processed for determination of glucose uptake, and GLUT4 mRNA and protein expression levels. The PGC-1α, AKT, p38MAPK, AS160, pAKT, pP38MAPK and pAS160 expression levels were quantitatively assessed too. We found that pharmacological activation of GALR2 enhanced energy expenditure, and increased GLUT4 expression and translocation in skeletal muscle of mice during high-fat diet regimens. Activation of GALR2 alleviated insulin resistance through P38MAPK/PGC-1α/GLUT4 and AKT/AS160/GLUT4 pathway in the skeletal muscle of mice. Overall, these results identify that GALR2 is a regulator of insulin resistance and activation of GALR2 represents a promising strategy against obesity-induced insulin resistance.


Subject(s)
Galanin/analogs & derivatives , Insulin Resistance , Muscle Proteins , Muscle, Skeletal/metabolism , Obesity/metabolism , Receptor, Galanin, Type 2 , Animals , Galanin/pharmacology , Glucose/metabolism , Male , Mice , Muscle Proteins/agonists , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Obesity/drug therapy , Obesity/pathology , Receptor, Galanin, Type 2/agonists , Receptor, Galanin, Type 2/metabolism
7.
J Biol Chem ; 292(34): 14270-14278, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28659340

ABSTRACT

Smooth muscle cell (SMC) differentiation is essential for vascular development, and TGF-ß signaling plays a critical role in this process. Although long non-coding RNAs (lncRNAs) regulate various cellular events, their functions in SMC differentiation remain largely unknown. Here, we demonstrate that the lncRNA growth arrest-specific 5 (GAS5) suppresses TGF-ß/Smad3 signaling in smooth muscle cell differentiation of mesenchymal progenitor cells. We found that forced expression of GAS5 blocked, but knockdown of GAS5 increased, the expression of SMC contractile proteins. Mechanistically, GAS5 competitively bound Smad3 protein via multiple RNA Smad-binding elements (rSBEs), which prevented Smad3 from binding to SBE DNA in TGF-ß-responsive SMC gene promoters, resulting in suppression of SMC marker gene transcription and, consequently, in inhibition of TGF-ß/Smad3-mediated SMC differentiation. Importantly, other lncRNAs or artificially synthesized RNA molecules that contained rSBEs also effectively inhibited TGF-ß/Smad3 signaling, suggesting that lncRNA-rSBE may be a general mechanism used by cells to fine-tune Smad3 activity in both basal and TGF-ß-stimulated states. Taken together, our results have uncovered an lncRNA-based mechanism that modulates TGF-ß/Smad3 signaling during SMC differentiation.


Subject(s)
Muscle, Smooth, Vascular/metabolism , RNA, Long Noncoding/metabolism , RNA/metabolism , Response Elements , Signal Transduction , Smad3 Protein/antagonists & inhibitors , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Binding, Competitive , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Gene Expression Regulation, Developmental/drug effects , Genes, Reporter/drug effects , In Situ Hybridization, Fluorescence , Isoquinolines/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Muscle Proteins/agonists , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nucleic Acid Conformation , Pyridines/pharmacology , Pyrroles/pharmacology , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/chemistry , Response Elements/drug effects , Signal Transduction/drug effects , Smad3 Protein/chemistry , Smad3 Protein/metabolism , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
8.
Am J Physiol Endocrinol Metab ; 312(6): E495-E507, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28246104

ABSTRACT

Cushing's syndrome is caused by overproduction of the adrenocorticotropic hormone (ACTH), which stimulates the adrenal grand to make cortisol. Skeletal muscle wasting occurs in pathophysiological response to Cushing's syndrome. The forkhead box (FOX) protein family has been implicated as a key regulator of muscle loss under conditions such as diabetes and sepsis. However, the mechanistic role of the FOXO family in ACTH-induced muscle atrophy is not understood. We hypothesized that FOXO3a plays a role in muscle atrophy through expression of the E3 ubiquitin ligases, muscle RING finger protein-1 (MuRF-1), and atrogin-1 in Cushing's syndrome. For establishment of a Cushing's syndrome animal model, Sprague-Dawley rats were implanted with osmotic minipumps containing ACTH (40 ng·kg-1·day-1). ACTH infusion significantly reduced muscle weight. In ACTH-infused rats, MuRF-1, atrogin-1, and FOXO3a were upregulated and the FOXO3a promoter was targeted by the glucocorticoid receptor (GR). Transcriptional activity and expression of FOXO3a were significantly decreased by the GR antagonist RU486. Treatment with RU486 reduced MuRF-1 and atrogin-1 expression in accordance with reduced enrichment of FOXO3a and Pol II on the promoters. Knockdown of FOXO3a prevented dexamethasone-induced MuRF-1 and atrogin-1 expression. These results indicate that FOXO3a plays a role in muscle atrophy through expression of MuRF-1 and atrogin-1 in Cushing's syndrome.


Subject(s)
Cushing Syndrome/metabolism , Disease Models, Animal , Forkhead Box Protein O3/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Chromatin Immunoprecipitation , Cushing Syndrome/pathology , Cushing Syndrome/physiopathology , Forkhead Box Protein O3/agonists , Forkhead Box Protein O3/antagonists & inhibitors , Forkhead Box Protein O3/genetics , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Glucocorticoids/pharmacology , Hormone Antagonists/pharmacology , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Promoter Regions, Genetic/drug effects , RNA Interference , Rats, Sprague-Dawley , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Response Elements/drug effects , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins/agonists , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
9.
Matrix Biol ; 57-58: 106-123, 2017 01.
Article in English | MEDLINE | ID: mdl-27964993

ABSTRACT

Previous work has shown that myotubes cultured on laminin-coated substrates form complex aggregates of synaptic proteins that are similar in shape and composition to neuromuscular junctions (NMJs). Here we show that laminin instructs the location of complex aggregates which form only on the lower surface when laminin is coated onto culture dishes but over the entire cell when laminin is added in solution. Silencing of myotubes by agents that block electrical activity (tetrodotoxin, verapamil) or by inhibitors of calmodulin dependent kinase (CaMKII) render the myotube permissive for the formation of complex aggregates. Treatment with laminin alone will facilitate the formation of complex aggregates hours later when myotubes are made permissive by inhibiting CaMKII. The AChR agonist carbachol disperses pre formed aggregates suggesting that non-permissiveness may involve active dispersal of AChRs. The permissive period requires ongoing protein synthesis. The latter may reflect a requirement for rapsyn, which turns over rapidly, and is necessary for aggregation. Consistent with this geldanamycin, an agent that increases rapsyn turnover disrupts complex aggregates. Agrin is well known to induce small clusters of AChRs but does not induce complex aggregates even though aggregate formation requires MuSK, a receptor tyrosine kinase activated by agrin. Dystroglycan (DG) is the major laminin receptor mediating complex aggregate formation with some contribution from ß1 integrins. In addition, there is a pool of CaMKII associated with DG. We discuss how these permissive and instructive mechanisms bear on NMJ formation in vivo.


Subject(s)
Calcium Channels/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Laminin/pharmacology , Muscle Fibers, Skeletal/drug effects , Protein Aggregates/drug effects , Receptors, Cholinergic/metabolism , Sodium Channels/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Agrin/genetics , Agrin/metabolism , Animals , Benzoquinones/pharmacology , Bungarotoxins/chemistry , Bungarotoxins/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Carbachol/pharmacology , Cell Line , Gene Expression , Lactams, Macrocyclic/pharmacology , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/agonists , Muscle Proteins/genetics , Muscle Proteins/metabolism , Receptors, Cholinergic/genetics , Sodium Channel Blockers/pharmacology , Sodium Channels/genetics , Staining and Labeling/methods , Tetrodotoxin/pharmacology , Verapamil/pharmacology
10.
Mol Pharmacol ; 89(5): 575-84, 2016 May.
Article in English | MEDLINE | ID: mdl-26976945

ABSTRACT

Positive allosteric modulators (PAMs) of nicotinic acetylcholine (ACh) receptors (nAChRs) have potential clinical applications in the treatment of nicotine dependence and many neuropsychiatric conditions associated with decreased brain cholinergic activity, and 3-(2-chlorophenyl)-5-(5-methyl-1-(piperidin-4-yl)-1H-pyrrazol-4-yl)isoxazole (CMPI) has been identified as a PAM selective for neuronal nAChRs containing theα4 subunit. In this report, we compare CMPI interactions with low-sensitivity (α4)3(ß2)2 and high-sensitivity (α4)2(ß2)3 nAChRs, and with muscle-type nAChRs. In addition, we use the intrinsic reactivity of [(3)H]CMPI upon photolysis at 312 nm to identify its binding sites inTorpedonAChRs. Recording fromXenopusoocytes, we found that CMPI potentiated maximally the responses of (α4)3(ß2)2nAChR to 10µM ACh (EC10) by 400% and with anEC50of ∼1µM. CMPI produced a left shift of the ACh concentration-response curve without altering ACh efficacy. In contrast, CMPI inhibited (∼35% at 10µM) ACh responses of (α4)2(ß2)3nAChRs and fully inhibited human muscle andTorpedonAChRs with IC50values of ∼0.5µM. Upon irradiation at 312 nm, [(3)H]CMPI photoincorporated into eachTorpedo[(α1)2ß1γδ] nAChR subunit. Sequencing of peptide fragments isolated from [(3)H]CMPI-photolabeled nAChR subunits established photolabeling of amino acids contributing to the ACh binding sites (αTyr(190),αTyr(198),γTrp(55),γTyr(111),γTyr(117),δTrp(57)) that was fully inhibitable by agonist and lower-efficiency, state-dependent [(3)H]CMPI photolabeling within the ion channel. Our results establish that CMPI is a potent potentiator of nAChRs containing anα4:α4 subunit interface, and that its intrinsic photoreactivy makes it of potential use to identify its binding sites in the (α4)3(ß2)2nAChR.


Subject(s)
Models, Molecular , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Allosteric Regulation/drug effects , Animals , Binding Sites , Humans , Isoxazoles/pharmacology , Kinetics , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Photolysis , Protein Conformation , Protein Isoforms/agonists , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Pyrazoles/pharmacology , Radioligand Assay , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Torpedo , Xenopus laevis
11.
Curr Opin Clin Nutr Metab Care ; 19(3): 177-81, 2016 May.
Article in English | MEDLINE | ID: mdl-26870889

ABSTRACT

PURPOSE OF REVIEW: Denervation is a hallmark of age-related and other types of muscle wasting. This review focuses on recent insights and current viewpoints regarding the mechanisms and clinical relevance of maintaining the neuromuscular junction to counteract muscle wasting resulting from aging or neural disease/damage. RECENT FINDINGS: Activity-dependent regulation of autophagy, the agrin-muscle specific kinase-Lrp4 signaling axis, and sympathetic modulation are principal mechanisms involved in stabilizing the neuromuscular junction. These findings are derived from several animal models and were largely confirmed by human gene expression analysis as well as insights from rare neuromuscular diseases such as amyotrophic lateral sclerosis and congenital myasthenic syndromes. Based on these insights, agrin-derived fragments are currently being evaluated as biomarkers for age-related muscle wasting. Tuning of autophagy, of the agrin pathway, and of sympathetic input are being studied as clinical treatment of muscle wasting disorders. SUMMARY: Basic research has revealed that maintenance of neuromuscular junctions and a few signaling pathways are important in the context of age-dependent and other forms of muscle wasting. These findings have recently started to enter clinical practice, but further research needs to substantiate and refine our knowledge.


Subject(s)
Models, Biological , Muscular Atrophy/etiology , Nerve Degeneration/etiology , Neuromuscular Junction Diseases/etiology , Neuromuscular Junction/physiopathology , Wasting Syndrome/physiopathology , Animals , Autophagy , Gene Expression Regulation , Humans , Muscle Proteins/agonists , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Wasting Syndrome/metabolism , Wasting Syndrome/pathology , Wnt Signaling Pathway
12.
PLoS One ; 10(12): e0144550, 2015.
Article in English | MEDLINE | ID: mdl-26658258

ABSTRACT

One of the causes of permanent disability in chronic multiple sclerosis patients is the inability of oligodendrocyte progenitor cells (OPCs) to terminate their maturation program at lesions. To identify key regulators of myelin gene expression acting at the last stages of OPC maturation we developed a drug repositioning strategy based on the mouse immortalized oligodendrocyte (OL) cell line Oli-neu brought to the premyelination stage by stably expressing a key factor regulating the last stages of OL maturation. The Prestwick Chemical Library of 1,200 FDA-approved compound(s) was repositioned at three dosages based on the induction of Myelin Basic Protein (MBP) expression. Drug hits were further validated using dosage-dependent reproducibility tests and biochemical assays. The glucocorticoid class of compounds was the most highly represented and we found that they can be divided in three groups according to their efficacy on MBP up-regulation. Since target identification is crucial before bringing compounds to the clinic, we searched for common targets of the primary screen hits based on their known chemical-target interactomes, and the pathways predicted by top ranking compounds were validated using specific inhibitors. Two of the top ranking compounds, Halcinonide and Clobetasol, act as Smoothened (Smo) agonists to up-regulate myelin gene expression in the Oli-neuM cell line. Further, RxRγ activation is required for MBP expression upon Halcinonide and Clobetasol treatment. These data indicate Clobetasol and Halcinonide as potential promyelinating drugs and also provide a mechanistic understanding of their mode of action in the pathway leading to myelination in OPCs. Furthermore, our classification of glucocorticoids with respect to MBP expression provides important novel insights into their effects in the CNS and a rational criteria for their choice in combinatorial therapies in de-myelinating diseases.


Subject(s)
Clobetasol/pharmacology , Cytoskeletal Proteins/metabolism , Halcinonide/pharmacology , Muscle Proteins/metabolism , Myelin Sheath/metabolism , Retinoid X Receptor gamma/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Cytoskeletal Proteins/agonists , Drug Repositioning , Gene Expression/drug effects , Immunoblotting , Mice , Microscopy, Fluorescence , Muscle Proteins/agonists , Myelin Basic Protein/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Retinoid X Receptor gamma/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
13.
FASEB J ; 29(11): 4544-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26178166

ABSTRACT

Early treatment with heart failure drugs lisinopril and spironolactone improves skeletal muscle pathology in Duchenne muscular dystrophy (DMD) mouse models. The angiotensin converting enzyme inhibitor lisinopril and mineralocorticoid receptor (MR) antagonist spironolactone indirectly and directly target MR. The presence and function of MR in skeletal muscle have not been explored. MR mRNA and protein are present in all tested skeletal muscles from both wild-type mice and DMD mouse models. MR expression is cell autonomous in both undifferentiated myoblasts and differentiated myotubes from mouse and human skeletal muscle cultures. To test for MR function in skeletal muscle, global gene expression analysis was conducted on human myotubes treated with MR agonist (aldosterone; EC50 1.3 nM) or antagonist (spironolactone; IC50 1.6 nM), and 53 gene expression differences were identified. Five differences were conserved in quadriceps muscles from dystrophic mice treated with spironolactone plus lisinopril (IC50 0.1 nM) compared with untreated controls. Genes down-regulated more than 2-fold by MR antagonism included FOS, ANKRD1, and GADD45B, with known roles in skeletal muscle, in addition to NPR3 and SERPINA3, bona fide targets of MR in other tissues. MR is a novel drug target in skeletal muscle and use of clinically safe antagonists may be beneficial for muscle diseases.


Subject(s)
Aldosterone/pharmacology , Lisinopril/pharmacology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins , Receptors, Melanocortin , Spironolactone/pharmacology , Animals , Cell Line , Humans , Mice , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/metabolism , Muscular Diseases/drug therapy , Muscular Diseases/metabolism , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/metabolism
14.
Biochem Biophys Res Commun ; 464(1): 208-13, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26102030

ABSTRACT

Transcription factors of the nuclear factor-kappa B (NF-κB) family play a pivotal role in inflammation, immunity and cell survival responses. Recent studies revealed that NF-κB also regulates the processes of muscle atrophy. NF-κB activity is regulated by various factors, including ankyrin repeat domain 2 (AnkrD2), which belongs to the muscle ankyrin repeat protein family. Another member of this family, AnkrD1 is also a transcriptional effector. The expression levels of AnkrD1 are highly upregulated in denervated skeletal muscle, suggesting an involvement of AnkrD1 in NF-κB mediated cellular responses to paralysis. However, the molecular mechanism underlying the interactive role of AnkrD1 in NF-κB mediated cellular responses is not well understood. In the current study, we examined the effect of AnkrD1 on NF-κB activity and determined the interactions between AnkrD1 expression and NF-κB signaling induced by TNFα in differentiating C2C12 myoblasts. TNFα upregulated AnkrD1 mRNA and protein levels. AnkrD1-siRNA significantly increased TNFα-induced transcriptional activation of NF-κB, whereas overexpression of AnkrD1 inhibited TNFα-induced NF-κB activity. Co-immunoprecipitation studies demonstrated that AnkrD1 was able to bind p50 subunit of NF-κB and vice versa. Finally, CHIP assays revealed that AnkrD1 bound chromatin at a NF-κB binding site in the AnrkD2 promoter and required NF-κB to do so. These results provide evidence of signaling integration between AnkrD1 and NF-κB pathways, and suggest a novel anti-inflammatory role of AnkrD1 through feedback inhibition of NF-κB transcriptional activity by which AnkrD1 modulates the balance between physiological and pathological inflammatory responses in skeletal muscle.


Subject(s)
Muscle Proteins/genetics , Myoblasts/metabolism , NF-kappa B/genetics , Nuclear Proteins/genetics , Protein Subunits/genetics , RNA, Messenger/genetics , Repressor Proteins/genetics , Animals , Binding Sites , Cell Differentiation , Cell Line , Cell Survival , Chromatin/metabolism , Feedback, Physiological , Gene Expression Regulation , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts/drug effects , Myoblasts/pathology , NF-kappa B/metabolism , Nuclear Proteins/agonists , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Binding , Protein Subunits/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/agonists , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
15.
Adv Exp Med Biol ; 832: 1-8, 2015.
Article in English | MEDLINE | ID: mdl-25315626

ABSTRACT

Oxidative stress and inflammation play an important role in the catabolism of skeletal muscles. Recently, cigarette smoke (CS) was shown to stimulate muscle catabolism by activation of p38 MAPK and up-regulation of the muscle-specific E3 ubiquitin ligases (E3s) atrogin-1 and MuRF1 which are over-expressed during muscle atrophy. Peroxynitrite (ONOO-), an oxidative ingredient of CS, also produced during oxidative stress and inflammation, was previously shown to induce ubiquitination and degradation of muscle proteins. To investigate the involvement of p38 MAPK and the muscle-specific E3s in ONOO--induced muscle catabolism, C2 myotubes, differentiated from a myoblast cell line, were exposed to ONOO- (25 µM) in a time-dependent manner. Following exposure, degradation of myosin heavy chain (MyHC) and actin, activation of p38 MAPK, and levels of atrogin-1 and MuRF1 were studied by Western blotting. Peak phosphorylation of p38 MAPK was observed at 1 h of ONOO- exposure. ONOO- caused a significant increase in the levels of atrogin-1 and MuRF1. In accordance, a significant decrease in MyHC levels was observed in a time-dependent manner. These findings support previous studies in which the catabolic effects of ONOO- were shown. In addition, ONOO- was demonstrated to induce degradation of muscle proteins by activation of p38 MAPK and up-regulation of the muscle-specific E3s atrogin-1 and MuRF1.


Subject(s)
Muscle Fibers, Skeletal/drug effects , Muscle Proteins/metabolism , Myosin Heavy Chains/metabolism , Peroxynitrous Acid/pharmacology , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Line , Gene Expression Regulation , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/agonists , Muscle Proteins/genetics , Myosin Heavy Chains/antagonists & inhibitors , Myosin Heavy Chains/genetics , Phosphorylation , Proteolysis , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , p38 Mitogen-Activated Protein Kinases/genetics
16.
PLoS One ; 9(9): e109098, 2014.
Article in English | MEDLINE | ID: mdl-25268515

ABSTRACT

Our aim was to compare and combine 3 nutritional strategies to slow down the age-related loss of muscle mass in healthy old rats: 1) increase protein intake, which is likely to stimulate muscle protein anabolism; 2) use leucine rich, rapidly digested whey proteins as protein source (whey proteins are recognized as the most effective proteins to stimulate muscle protein anabolism). 3) Supplement animals with a mixture of chamomile extract, vitamin E, vitamin D (reducing inflammation and oxidative stress is also effective to improve muscle anabolism). Such comparisons and combinations were never tested before. Nutritional groups were: casein 12% protein, whey 12% protein, whey 18% protein and each of these groups were supplemented or not with polyphenols/antioxidants. During 6 months, we followed changes of weight, food intake, inflammation (plasma fibrinogen and alpha-2-macroglobulin) and body composition (DXA). After 6 months, we measured muscle mass, in vivo and ex-vivo fed and post-absorptive muscle protein synthesis, ex-vivo muscle proteolysis, and oxidative stress parameters (liver and muscle glutathione, SOD and total antioxidant activities, muscle carbonyls and TBARS). We showed that although micronutrient supplementation reduced inflammation and oxidative stress, the only factor that significantly reduced the loss of lean body mass was the increase in whey protein intake, with no detectable effect on muscle protein synthesis, and a tendency to reduce muscle proteolysis. We conclude that in healthy rats, increasing protein intake is an effective way to delay sarcopenia.


Subject(s)
Aging/drug effects , Dietary Supplements , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Sarcopenia/prevention & control , Whey Proteins/administration & dosage , Aging/metabolism , Animals , Body Weight/drug effects , Chamomile/chemistry , Diet/methods , Eating/drug effects , Fibrinogen/metabolism , Glutathione/metabolism , Male , Muscle Proteins/agonists , Muscle, Skeletal/metabolism , Organ Size/drug effects , Plant Extracts/administration & dosage , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Vitamin D/administration & dosage , Vitamin E/administration & dosage , alpha-Macroglobulins/metabolism
17.
Diabetes Obes Metab ; 16(8): 711-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24476050

ABSTRACT

AIMS: This work explored the effects of irisin on metabolism, gene expression and mitochondrial content in cultured myocytes. METHODS: C2C12 myocytes were treated with various concentrations of irisin for various durations. Glycolysis and oxidative metabolism were quantified by measurement of extracellular acidification and oxygen consumption, respectively. Metabolic gene expression was measured by quantitative real-time polymerase chain reaction (qRT-PCR) and mitochondrial content was assessed by flow cytometry and confocal microscopy. RESULTS: Cells treated with irisin exhibited significantly increased oxidative metabolism. Irisin treatment also significantly increased mitochondrial uncoupling at various doses and durations. Lastly, treatment with irisin also significantly elevated metabolic gene expression including peroxisome proliferator-activated receptor γ coactivator-1 alpha (PGC-1α), nuclear respiratory factor 1 (NRF1), mitochondrial transcription factor A (TFAM), irisin, glucose transporter 4 (GLUT4) and mitochondrial uncoupling protein 3 (UCP3) leading to increased mitochondrial biogenesis. CONCLUSIONS: Our observations are the first to document increased metabolism in myocytes through irisin-mediated induction of mitochondrial biogenesis and uncoupling with corresponding gene expression. These observations support the need for further investigation into the therapeutic and pharmacological effects of irisin, as well as development of irisin-based therapy.


Subject(s)
Fibronectins/pharmacology , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Mitochondria, Muscle/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/metabolism , Oxidative Phosphorylation/drug effects , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA-Binding Proteins/agonists , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibronectins/agonists , Fibronectins/genetics , Fibronectins/metabolism , High Mobility Group Proteins/agonists , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , Kinetics , Mice , Mitochondria, Muscle/metabolism , Mitochondrial Turnover/drug effects , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/agonists , Muscle Proteins/genetics , Nuclear Respiratory Factor 1/agonists , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Recombinant Proteins/pharmacology , Transcription Factors/agonists , Transcription Factors/genetics , Transcription Factors/metabolism
18.
J Cell Sci ; 127(Pt 1): 40-9, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24155330

ABSTRACT

Cardiac fibrosis is linked to fibroblast-to-myofibroblast phenoconversion and proliferation but the mechanisms underlying this are poorly understood. Ski is a negative regulator of TGF-ß-Smad signaling in myofibroblasts, and might redirect the myofibroblast phenotype back to fibroblasts. Meox2 could alter TGF-ß-mediated cellular processes and is repressed by Zeb2. Here, we investigated whether Ski diminishes the myofibroblast phenotype by de-repressing Meox2 expression and function through repression of Zeb2 expression. We show that expression of Meox1 and Meox2 mRNA and Meox2 protein is reduced during phenoconversion of fibroblasts to myofibroblasts. Overexpression of Meox2 shifts the myofibroblasts into fibroblasts, whereas the Meox2 DNA-binding mutant has no effect on myofibroblast phenotype. Overexpression of Ski partially restores Meox2 mRNA expression levels to those in cardiac fibroblasts. Expression of Zeb2 increased during phenoconversion and Ski overexpression reduces Zeb2 expression in first-passage myofibroblasts. Furthermore, expression of Meox2 is decreased in scar following myocardial infarction, whereas Zeb2 protein expression increases in the infarct scar. Thus Ski modulates the cardiac myofibroblast phenotype and function through suppression of Zeb2 by upregulating the expression of Meox2. This cascade might regulate cardiac myofibroblast phenotype and presents therapeutic options for treatment of cardiac fibrosis.


Subject(s)
Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Myofibroblasts/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Fibroblasts/pathology , Fibrosis , Gene Expression Regulation , Homeodomain Proteins/agonists , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Muscle Proteins/agonists , Muscle Proteins/genetics , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Myofibroblasts/pathology , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
19.
Biochemistry ; 52(47): 8480-8, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24164482

ABSTRACT

The differential action of the novel agonist JN403 at neuronal α7 and muscle nicotinic receptors (AChRs) was explored by using a combination of functional and structural approaches. Single-channel recordings reveal that JN403 is a potent agonist of α7 but a very low-efficacy agonist of muscle AChRs. JN403 elicits detectable openings of α7 and muscle AChRs at concentrations ~1000-fold lower and ~20-fold higher, respectively, than that for ACh. Single-channel activity elicited by JN403 is very similar to that elicited by ACh in α7 but profoundly different in muscle AChRs, where openings are brief and infrequent and do not appear in clusters at any concentration. JN403 elicits single-channel activity of muscle AChRs lacking the ε subunit, with opening events being more frequent and prolonged than those of wild-type AChRs. This finding is in line with the molecular docking studies predicting that JN403 may form a hydrogen bond required for potent activation at the α-δ but not at the α-ε binding site. JN403 does not elicit detectable Ca²âº influx in muscle AChRs but inhibits (±)-epibatidine-elicited influx mainly by a noncompetitive mechanism. Such inhibition is compatible with single-channel recordings revealing that JN403 produces open-channel blockade and early termination of ACh-elicited clusters, and it is therefore also a potent desensitizing enhancer of muscle AChRs. The latter mechanism is supported by the JN403-induced increase in the level of binding of [³H]cytisine and [³H]TCP to resting AChRs. Elucidation of the differences in activity of JN403 between neuronal α7 and muscle AChRs provides further insights into mechanisms underlying selectivity for α7 AChRs.


Subject(s)
Carbamates/pharmacology , Muscle Proteins/agonists , Nerve Tissue Proteins/agonists , Nicotinic Agonists/pharmacology , Quinuclidines/pharmacology , Receptors, Nicotinic/metabolism , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Calcium Signaling/drug effects , Carbamates/metabolism , Cell Line , Fetal Proteins/agonists , Fetal Proteins/chemistry , Fetal Proteins/genetics , Fetal Proteins/metabolism , Humans , Kinetics , Membrane Potentials/drug effects , Mice , Molecular Conformation , Molecular Docking Simulation , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nicotinic Agonists/metabolism , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/metabolism , Nicotinic Antagonists/pharmacology , Protein Binding , Protein Subunits/agonists , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Quinuclidines/metabolism , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Torpedo , alpha7 Nicotinic Acetylcholine Receptor/chemistry , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
20.
J Cardiovasc Pharmacol ; 62(6): 497-506, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24084216

ABSTRACT

Cardinal pathological features of hypertensive heart disease (HHD) include not only hypertrophied cardiomyocytes and foci of scattered microscopic scarring, a footprint of prior necrosis, but also small myocytes ensnared by fibrillar collagen where disuse atrophy with protein degradation would be predicted. Whether atrophic signaling is concordant with the appearance of HHD and involves oxidative and endoplasmic reticulum (ER) stress remains unexplored. Herein, we examine these possibilities focusing on the left ventricle and cardiomyocytes harvested from hypertensive rats receiving 4 weeks aldosterone/salt treatment (ALDOST) alone or together with ZnSO4, a nonvasoactive antioxidant, with the potential to attenuate atrophy and optimize hypertrophy. Compared with untreated age-/sex-/strain-matched controls, ALDOST was accompanied by (1) left ventricle hypertrophy with preserved systolic function; (2) concordant cardiomyocyte atrophy (<1000 µm²) found at sites bordering on fibrosis where they were reexpressing ß-myosin heavy chain; and (3) upregulation of ubiquitin ligases, muscle RING-finger protein-1 and atrogin-1, and elevated 8-isoprostane and unfolded protein ER response with messenger RNA upregulation of stress markers. ZnSO4 cotreatment reduced lipid peroxidation, fibrosis, and the number of atrophic myocytes, together with a further increase in cell area and width of atrophied and hypertrophied myocytes, and improved systolic function but did not attenuate elevated blood pressure. We conclude that atrophic signaling, concordant with hypertrophy, occurs in the presence of a reparative fibrosis and induction of oxidative and ER stress at sites of scarring where myocytes are atrophied. ZnSO4 cotreatment in HHD with ALDOST attenuates the number of atrophic myocytes, optimizes size of atrophied and hypertrophied myocytes, and improves systolic function.


Subject(s)
Disease Models, Animal , Hypertension/metabolism , Hypertrophy, Left Ventricular/etiology , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cell Size/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Hypertension/drug therapy , Hypertension/pathology , Hypertension/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Male , Muscle Proteins/agonists , Muscle Proteins/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction/drug effects , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL