Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
NPJ Microgravity ; 9(1): 2, 2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36646717

ABSTRACT

Novel treatments for muscle wasting are of significant value to patients with disease states that result in muscle weakness, injury recovery after immobilization and bed rest, and for astronauts participating in long-duration spaceflight. We utilized an anti-myostatin peptibody to evaluate how myostatin signaling contributes to muscle loss in hindlimb suspension. Male C57BL/6 mice were left non-suspended (NS) or were hindlimb suspended (HS) for 14 days and treated with a placebo vehicle (P) or anti-myostatin peptibody (D). Hindlimb suspension (HS-P) resulted in rapid and significantly decreased body mass (-5.6% by day 13) with hindlimb skeletal muscle mass losses between -11.2% and -22.5% and treatment with myostatin inhibitor (HS-D) partially attenuated these losses. Myostatin inhibition increased hindlimb strength with no effect on soleus tetanic strength. Soleus mass and fiber CSA were reduced with suspension and did not increase with myostatin inhibition. In contrast, the gastrocnemius showed histological evidence of wasting with suspension that was partially mitigated with myostatin inhibition. While expression of genes related to protein degradation (Atrogin-1 and Murf-1) in the tibialis anterior increased with suspension, these atrogenes were not significantly reduced by myostatin inhibition despite a modest activation of the Akt/mTOR pathway. Taken together, these findings suggest that myostatin is important in hindlimb suspension but also motivates the study of other factors that contribute to disuse muscle wasting. Myostatin inhibition benefitted skeletal muscle size and function, which suggests therapeutic potential for both spaceflight and terrestrial applications.

2.
J Muscle Res Cell Motil ; 44(2): 95-106, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36316565

ABSTRACT

Non-traditional animal models present an opportunity to discover novel biology that has evolved to allow such animals to survive in extreme environments. One striking example is the Burmese python (Python molurus bivittatus), which exhibits extreme physiological adaptation in various metabolic organs after consuming a large meal following long periods of fasting. The response to such a large meal in pythons involves a dramatic surge in metabolic rate, lipid overload in plasma, and massive but reversible organ growth through the course of digestion. Multiple studies have reported the physiological responses in post-prandial pythons, while the specific molecular control of these processes is less well-studied. Investigating the mechanisms that coordinate organ growth and adaptive responses offers the opportunity to gain novel insight that may be able to treat various pathologies in humans. Here, we summarize past research on the post-prandial physiological changes in the Burmese python with a focus on the gastrointestinal tract, heart, and liver. Specifically, we address our recent molecular discoveries in the post-prandial python liver which demonstrate transient adaptations that may reveal new therapeutic targets. Lastly, we explore new biology of the aquaporin 7 gene that is potently upregulated in mammalian cardiac myocytes by circulating factors in post-prandial python plasma.


Subject(s)
Boidae , Postprandial Period , Animals , Boidae/genetics , Boidae/metabolism , Boidae/physiology , Mammals , Myanmar , Postprandial Period/physiology
3.
J Gen Physiol ; 154(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35323838

ABSTRACT

As an opportunistic predator, the Burmese python (Python molurus bivittatus) consumes large and infrequent meals, fasting for up to a year. Upon consuming a large meal, the Burmese python exhibits extreme metabolic responses. To define the pathways that regulate these postprandial metabolic responses, we performed a comprehensive profile of plasma metabolites throughout the digestive process. Following ingestion of a meal equivalent to 25% of its body mass, plasma lipoproteins and metabolites, such as chylomicra and bile acids, reach levels observed only in mammalian models of extreme dyslipidemia. Here, we provide evidence for an adaptive response to postprandial nutrient overload by the python liver, a critical site of metabolic homeostasis. The python liver undergoes a substantial increase in mass through proliferative processes, exhibits hepatic steatosis, hyperlipidemia-induced insulin resistance indicated by PEPCK activation and pAKT deactivation, and de novo fatty acid synthesis via FASN activation. This postprandial state is completely reversible. We posit that Burmese pythons evade the permanent hepatic damage associated with these metabolic states in mammals using evolved protective measures to inactivate these pathways. These include a transient activation of hepatic nuclear receptors induced by fatty acids and bile acids, including PPAR and FXR, respectively. The stress-induced p38 MAPK pathway is also transiently activated during the early stages of digestion. Taken together, these data identify a reversible metabolic response to hyperlipidemia by the python liver, only achieved in mammals by pharmacologic intervention. The factors involved in these processes may be relevant to or leveraged for remediating human hepatic pathology.


Subject(s)
Boidae , Adaptation, Physiological , Animals , Boidae/metabolism , Humans , Liver , Mammals , Nutrients , Postprandial Period/physiology
4.
J Cell Sci ; 133(15)2020 08 11.
Article in English | MEDLINE | ID: mdl-32620696

ABSTRACT

Striated muscle is a highly specialized collection of tissues with contractile properties that vary according to functional needs. Although muscle fiber types are established postnatally, lifelong plasticity facilitates stimulus-dependent adaptation. Functional adaptation requires molecular adaptation, which is partially provided by miRNA-mediated post-transcriptional regulation. miR-206 is a muscle-specific miRNA enriched in slow muscles. We investigated whether miR-206 drives the slow muscle phenotype or is merely an outcome. We found that miR-206 expression increases in both physiological (including female sex and endurance exercise) and pathological conditions (muscular dystrophy and adrenergic agonism) that promote a slow phenotype. Consistent with that observation, the slow soleus muscle of male miR-206-knockout mice displays a faster phenotype than wild-type mice. Moreover, left ventricles of male miR-206 knockout mice have a faster myosin profile, accompanied by dilation and systolic dysfunction. Thus, miR-206 appears to be necessary to enforce a slow skeletal and cardiac muscle phenotype and to play a key role in muscle sexual dimorphisms.


Subject(s)
MicroRNAs , Muscle, Skeletal , Animals , Female , Male , Mice , MicroRNAs/genetics , Muscle Contraction/genetics , Muscle Fibers, Skeletal , Phenotype
5.
Science ; 351(6273): 617-21, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26912705

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is an inherited disease of heart muscle that can be caused by mutations in sarcomere proteins. Clinical diagnosis depends on an abnormal thickening of the heart, but the earliest signs of disease are hyperdynamic contraction and impaired relaxation. Whereas some in vitro studies of power generation by mutant and wild-type sarcomere proteins are consistent with mutant sarcomeres exhibiting enhanced contractile power, others are not. We identified a small molecule, MYK-461, that reduces contractility by decreasing the adenosine triphosphatase activity of the cardiac myosin heavy chain. Here we demonstrate that early, chronic administration of MYK-461 suppresses the development of ventricular hypertrophy, cardiomyocyte disarray, and myocardial fibrosis and attenuates hypertrophic and profibrotic gene expression in mice harboring heterozygous human mutations in the myosin heavy chain. These data indicate that hyperdynamic contraction is essential for HCM pathobiology and that inhibitors of sarcomere contraction may be a valuable therapeutic approach for HCM.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Benzylamines/administration & dosage , Cardiac Myosins/antagonists & inhibitors , Cardiomyopathy, Hypertrophic, Familial/drug therapy , Myocardial Contraction/drug effects , Myosin Heavy Chains/antagonists & inhibitors , Sarcomeres/drug effects , Uracil/analogs & derivatives , Animals , Benzylamines/chemistry , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic, Familial/pathology , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Cells, Cultured , Disease Models, Animal , Fibrosis , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heterozygote , Humans , Male , Mice , Mice, Inbred Strains , Mutation , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Rats , Uracil/administration & dosage , Uracil/chemistry
6.
FEBS Lett ; 589(10): 1080-8, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25816750

ABSTRACT

PKD-mediated phosphorylation of class IIa HDACs frees the MEF2 transcription factor to activate genes that govern muscle differentiation and growth. Studies of the regulation and function of this signaling axis have involved MC1568 and Gö-6976, which are small molecule inhibitors of class IIa HDAC and PKD catalytic activity, respectively. We describe unanticipated effects of these compounds. MC1568 failed to inhibit class IIa HDAC catalytic activity in vitro, and exerted divergent effects on skeletal muscle differentiation compared to a bona fide inhibitor of these HDACs. In cardiomyocytes, Gö-6976 triggered calcium signaling and activated stress-inducible kinases. Based on these findings, caution is warranted when employing MC1568 and Gö-6976 as pharmacological tool compounds to assess functions of class IIa HDACs and PKD.


Subject(s)
Carbazoles/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Muscle Proteins/antagonists & inhibitors , Muscle, Skeletal/metabolism , Protein Kinase C/antagonists & inhibitors , Pyrroles/pharmacology , Animals , Calcium Signaling/drug effects , Carbazoles/chemistry , Cell Line , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/genetics , Hydroxamic Acids/chemistry , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Pyrroles/chemistry
7.
PLoS One ; 10(2): e0118229, 2015.
Article in English | MEDLINE | ID: mdl-25689854

ABSTRACT

microRNAs (miRNAs) are short non-coding RNAs that can mediate changes in gene expression and are required for the formation of skeletal muscle (myogenesis). With the goal of identifying novel miRNA biomarkers of muscle disease, we profiled miRNA expression using miRNA-seq in the gastrocnemius muscles of dystrophic mdx4cv mice. After identifying a down-regulation of the miR-30 family (miR-30a-5p, -30b, -30c, -30d and -30e) when compared to C57Bl/6 (WT) mice, we found that overexpression of miR-30 family miRNAs promotes differentiation, while inhibition restricts differentiation of myoblasts in vitro. Additionally, miR-30 family miRNAs are coordinately down-regulated during in vivo models of muscle injury (barium chloride injection) and muscle disuse atrophy (hindlimb suspension). Using bioinformatics tools and in vitro studies, we identified and validated Smarcd2, Snai2 and Tnrc6a as miR-30 family targets. Interestingly, we show that by targeting Tnrc6a, miR-30 family miRNAs negatively regulate the miRNA pathway and modulate both the activity of muscle-specific miR-206 and the levels of protein synthesis. These findings indicate that the miR-30 family may be an interesting biomarker of perturbed muscle homeostasis and muscle disease.


Subject(s)
Cell Differentiation/genetics , Feedback, Physiological , MicroRNAs/genetics , Muscle Development/genetics , Animals , Autoantigens/genetics , Chromosomal Proteins, Non-Histone/genetics , Epigenesis, Genetic , Humans , Male , Mice , Muscle Proteins/genetics , Muscles/cytology , Muscles/injuries , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription, Genetic
8.
Muscle Nerve ; 48(3): 393-402, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23893872

ABSTRACT

INTRODUCTION: Hindlimb unloading-induced muscle atrophy is often assessed after a homeostatic state is established, thus overlooking the early adaptations that are critical to developing this pattern of atrophy. METHODS: Muscle function and physiology were characterized at 0, 1, 3, 7, and 14 days of hindlimb suspension (HS). RESULTS: Reductions in muscle mass were maximal by Day 14 of HS. Functional strength and isolated muscle strength were reduced. MyHC-I and -IIa expressing fibers were reduced in size by Day 7 in the soleus and by Day 14 in the gastrocnemius (MyHC-I fibers only). Atrogin-1 and MuRF1 expression was increased by Day 1 in both the calf and tibialis anterior while IGF-1 expression was significantly reduced on Day 3. Phosphorylation of Akt was reduced on Day 14. CONCLUSIONS: Insight into these early changes in response to HS improves understanding of the molecular and functional changes that lead to muscle atrophy.


Subject(s)
Adaptation, Biological/physiology , Gene Expression Regulation/physiology , Hindlimb Suspension , Muscle, Skeletal/physiology , Action Potentials , Analysis of Variance , Animals , Body Mass Index , Electric Stimulation , Exercise Test , Insulin-Like Growth Factor I/metabolism , Longitudinal Studies , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Muscle Fatigue/physiology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscle Strength , Muscle, Skeletal/chemistry , Myosin Heavy Chains/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Time Factors , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism
9.
Proc Natl Acad Sci U S A ; 110(24): 9806-11, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23720316

ABSTRACT

Cardiac hypertrophy is a strong predictor of morbidity and mortality in patients with heart failure. Small molecule histone deacetylase (HDAC) inhibitors have been shown to suppress cardiac hypertrophy through mechanisms that remain poorly understood. We report that class I HDACs function as signal-dependent repressors of cardiac hypertrophy via inhibition of the gene encoding dual-specificity phosphatase 5 (DUSP5) DUSP5, a nuclear phosphatase that negatively regulates prohypertrophic signaling by ERK1/2. Inhibition of DUSP5 by class I HDACs requires activity of the ERK kinase, mitogen-activated protein kinase kinase (MEK), revealing a self-reinforcing mechanism for promotion of cardiac ERK signaling. In cardiac myocytes treated with highly selective class I HDAC inhibitors, nuclear ERK1/2 signaling is suppressed in a manner that is absolutely dependent on DUSP5. In contrast, cytosolic ERK1/2 activation is maintained under these same conditions. Ectopic expression of DUSP5 in cardiomyocytes results in potent inhibition of agonist-dependent hypertrophy through a mechanism involving suppression of the gene program for hypertrophic growth. These findings define unique roles for class I HDACs and DUSP5 as integral components of a regulatory signaling circuit that controls cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Dual-Specificity Phosphatases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Histone Deacetylases/metabolism , Animals , Animals, Newborn , Benzamides/pharmacology , Cardiomegaly/genetics , Cell Nucleus/enzymology , Cells, Cultured , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Immunoblotting , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pyrimidines/pharmacology , RNA Interference , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
10.
Cell ; 153(4): 743-5, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23663775

ABSTRACT

In this issue of Cell, Loffredo et al. demonstrate that exposing an old mouse to the circulatory system of a young mouse reverses age-related cardiac hypertrophy. The authors demonstrate that this effect can be recapitulated by treating old mice with growth and differentiation factor 11 (GDF11). These data suggest that GDF11 therapy may be a useful tool in combating age-related cardiac hypertrophy.

11.
Cell ; 151(6): 1319-31, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23217713

ABSTRACT

PGC-1α is a transcriptional coactivator induced by exercise that gives muscle many of the best known adaptations to endurance-type exercise but has no effects on muscle strength or hypertrophy. We have identified a form of PGC-1α (PGC-1α4) that results from alternative promoter usage and splicing of the primary transcript. PGC-1α4 is highly expressed in exercised muscle but does not regulate most known PGC-1α targets such as the mitochondrial OXPHOS genes. Rather, it specifically induces IGF1 and represses myostatin, and expression of PGC-1α4 in vitro and in vivo induces robust skeletal muscle hypertrophy. Importantly, mice with skeletal muscle-specific transgenic expression of PGC-1α4 show increased muscle mass and strength and dramatic resistance to the muscle wasting of cancer cachexia. Expression of PGC-1α4 is preferentially induced in mouse and human muscle during resistance exercise. These studies identify a PGC-1α protein that regulates and coordinates factors involved in skeletal muscle hypertrophy.


Subject(s)
Heat-Shock Proteins/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Resistance Training , Trans-Activators/metabolism , Transcription Factors/metabolism , Adiposity , Animals , Glucose/metabolism , Humans , Hypertrophy , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle Fibers, Skeletal/metabolism , Myostatin/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Isoforms/metabolism
12.
Science ; 334(6055): 528-31, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22034436

ABSTRACT

Burmese pythons display a marked increase in heart mass after a large meal. We investigated the molecular mechanisms of this physiological heart growth with the goal of applying this knowledge to the mammalian heart. We found that heart growth in pythons is characterized by myocyte hypertrophy in the absence of cell proliferation and by activation of physiological signal transduction pathways. Despite high levels of circulating lipids, the postprandial python heart does not accumulate triglycerides or fatty acids. Instead, there is robust activation of pathways of fatty acid transport and oxidation combined with increased expression and activity of superoxide dismutase, a cardioprotective enzyme. We also identified a combination of fatty acids in python plasma that promotes physiological heart growth when injected into either pythons or mice.


Subject(s)
Boidae/physiology , Fatty Acids/metabolism , Heart/growth & development , Animals , Animals, Newborn , Biological Transport , Boidae/anatomy & histology , Boidae/genetics , Cardiomegaly , Cell Size , Fasting , Fatty Acids/blood , Fatty Acids, Monounsaturated/blood , Fatty Acids, Monounsaturated/pharmacology , Fatty Acids, Nonesterified/blood , Female , Gene Expression Regulation , Heart/anatomy & histology , Heart/drug effects , Male , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myristic Acids/blood , Myristic Acids/pharmacology , Oxidation-Reduction , Palmitic Acid/blood , Palmitic Acid/pharmacology , Postprandial Period , Protein Biosynthesis , Superoxide Dismutase/metabolism , Triglycerides/blood
13.
Skelet Muscle ; 1(1): 5, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21798083

ABSTRACT

BACKGROUND: While the myosin heavy chain IIb isoform (MyHC-IIb) is the predominant motor protein in most skeletal muscles of rats and mice, the messenger RNA (mRNA) for this isoform is only expressed in a very small subset of specialized muscles in adult large mammals, including humans. RESULTS: We identify the DNA sequences limiting MyHC-IIb expression in humans and explore the activation of this gene in human skeletal muscle. We demonstrate that the transcriptional activity of ~1.0 kb of the human MyHC-IIb promoter is greatly reduced compared to that of the corresponding mouse sequence in both mouse and human myotubes in vitro and show that nucleotide differences that eliminate binding sites for myocyte enhancer factor 2 (MEF2) and serum response factor (SRF) account for this difference. Despite these differences, we show that MyHC-IIb mRNA is expressed in fetal human muscle cells and that MyHC-IIb mRNA is significantly up-regulated in the skeletal muscle of Duchene muscular dystrophy patients. CONCLUSIONS: These data identify the genetic basis for a key phenotypic difference between the muscles of large and small mammals, and demonstrate that mRNA expression of the MyHC-IIb gene can be re-activated in human limb muscle undergoing profound degeneration/regeneration.

14.
FEBS Lett ; 584(6): 1103-10, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20188095

ABSTRACT

Class IIa histone deacetylases (HDACs) -4, -5, -7 and -9 undergo signal-dependent nuclear export upon phosphorylation of conserved serine residues that are targets for 14-3-3 binding. Little is known of other mechanisms for regulating the subcellular distribution of class IIa HDACs. Using a biochemical purification strategy, we identified protein kinase C-related kinase-2 (PRK2) as an HDAC5-interacting protein. PRK2 and the related kinase, PRK1, phosphorylate HDAC5 at a threonine residue (Thr-292) positioned within the nuclear localization signal (NLS) of the protein. HDAC7 and HDAC9 contain analogous sites that are phosphorylated by PRK, while HDAC4 harbors a non-phosphorylatable alanine residue at this position. We provide evidence to suggest that the unique phospho-acceptor cooperates with the 14-3-3 target sites to impair HDAC nuclear import.


Subject(s)
Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Nuclear Localization Signals/metabolism , Protein Kinase C/metabolism , 14-3-3 Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Animals , COS Cells , Catalytic Domain , Cells, Cultured , Chlorocebus aethiops , Consensus Sequence , Humans , Models, Biological , Phosphorylation , Protein Binding , Protein Interaction Mapping
15.
J Am Coll Cardiol ; 54(21): 1971-81, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19909879

ABSTRACT

OBJECTIVES: This study evaluated the safety and efficacy of ambrisentan for a period of 2 years in patients with pulmonary arterial hypertension (PAH). BACKGROUND: Ambrisentan is an oral, once-daily endothelin receptor antagonist that is selective for the endothelin type A receptor. The ARIES-1 (Ambrisentan in Pulmonary Arterial Hypertension, Randomized, Double-Blind, Placebo-Controlled, Multicenter, Efficacy Studies) and ARIES-2 trials were the pivotal 12-week, placebo-controlled studies that led to the regulatory approval of ambrisentan (5 and 10 mg) for the treatment of PAH. METHODS: In the ARIES-1 and -2 studies, and the subsequent long-term extension protocol, the ARIES-E study, 383 patients received ambrisentan (2.5, 5, or 10 mg). Efficacy and safety assessments are presented from the time of the first dose of ambrisentan for all patients with post-baseline data. RESULTS: After 2 years of ambrisentan exposure, the mean change from baseline in 6-min walk distance was improved for the 5-mg (+23 m; 95% confidence interval: 9 to 38 m) and 10-mg (+28 m; 95% confidence interval: 11 to 45 m) groups. Estimates of survival and freedom from clinical worsening for the combined dose group were 94% and 83%, respectively, at 1 year and 88% and 72%, respectively, at 2 years. The annualized risk of aminotransferase abnormalities >3x the upper limit of normal was approximately 2% per year; most of these events were mild and did not lead to discontinuation of drug. CONCLUSIONS: Two years of ambrisentan treatment was associated with sustained improvements in exercise capacity and a low risk of clinical worsening and death in patients with PAH. Ambrisentan was generally well tolerated and had a low risk of aminotransferase abnormalities over the 2-year study period. (A Long Term Study of Ambrisentan in Pulmonary Arterial Hypertension Subjects Having Completed AMB-320 or AMB-321; NCT00578786).


Subject(s)
Hypertension, Pulmonary/drug therapy , Phenylpropionates/administration & dosage , Pyridazines/administration & dosage , Administration, Oral , Dose-Response Relationship, Drug , Double-Blind Method , Exercise Tolerance/drug effects , Female , Follow-Up Studies , Humans , Hypertension, Pulmonary/mortality , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Pulmonary Wedge Pressure/drug effects , Survival Rate/trends , Time Factors , Treatment Outcome
16.
J Appl Physiol (1985) ; 106(2): 582-95, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074574

ABSTRACT

Spaceflight results in a number of adaptations to skeletal muscle, including atrophy and shifts toward faster muscle fiber types. To identify changes in gene expression that may underlie these adaptations, we used both microarray expression analysis and real-time polymerase chain reaction to quantify shifts in mRNA levels in the gastrocnemius from mice flown on the 11-day, 19-h STS-108 shuttle flight and from normal gravity controls. Spaceflight data also were compared with the ground-based unloading model of hindlimb suspension, with one group of pure suspension and one of suspension followed by 3.5 h of reloading to mimic the time between landing and euthanization of the spaceflight mice. Analysis of microarray data revealed that 272 mRNAs were significantly altered by spaceflight, the majority of which displayed similar responses to hindlimb suspension, whereas reloading tended to counteract these responses. Several mRNAs altered by spaceflight were associated with muscle growth, including the phosphatidylinositol 3-kinase regulatory subunit p85alpha, insulin response substrate-1, the forkhead box O1 transcription factor, and MAFbx/atrogin1. Moreover, myostatin mRNA expression tended to increase, whereas mRNA levels of the myostatin inhibitor FSTL3 tended to decrease, in response to spaceflight. In addition, mRNA levels of the slow oxidative fiber-associated transcriptional coactivator peroxisome proliferator-associated receptor (PPAR)-gamma coactivator-1alpha and the transcription factor PPAR-alpha were significantly decreased in spaceflight gastrocnemius. Finally, spaceflight resulted in a significant decrease in levels of the microRNA miR-206. Together these data demonstrate that spaceflight induces significant changes in mRNA expression of genes associated with muscle growth and fiber type.


Subject(s)
Gene Expression Regulation , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Space Flight , Weightlessness , Adaptation, Physiological/genetics , Animals , Cluster Analysis , Female , Gene Expression Profiling/methods , Hindlimb Suspension , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology , Myostatin/genetics , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/metabolism , Reproducibility of Results , TOR Serine-Threonine Kinases , Time Factors
18.
Cell Metab ; 7(2): 97-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18249167

ABSTRACT

Akt1 is a well-characterized mediator of muscle hypertrophy. In this issue of Cell Metabolism, Izumiya et al. (2008) reveal a striking link between Akt1 signaling, fast muscle fiber size, and whole-body metabolism. These results provide new insights into the ability of muscle to combat diet-induced obesity and metabolic dysfunction.


Subject(s)
Adipose Tissue , Muscle, Skeletal/growth & development , Proto-Oncogene Proteins c-akt/physiology , Weight Loss/physiology , Animals , Female , Male , Muscle, Skeletal/physiology
19.
Mol Cell Biol ; 26(10): 3875-88, 2006 May.
Article in English | MEDLINE | ID: mdl-16648482

ABSTRACT

In response to pathological stresses such as hypertension or myocardial infarction, the heart undergoes a remodeling process that is associated with myocyte hypertrophy, myocyte death, and fibrosis. Histone deacetylase 5 (HDAC5) is a transcriptional repressor of cardiac remodeling that is subject to phosphorylation-dependent neutralization in response to stress signaling. Recent studies have suggested a role for protein kinase C (PKC) and its downstream effector, protein kinase D1 (PKD1), in the control of HDAC5 phosphorylation. While PKCs are well-documented regulators of cardiac signaling, the function of PKD1 in heart muscle remains unclear. Here, we demonstrate that PKD1 catalytic activity is stimulated in cardiac myocytes by diverse hypertrophic agonists that signal through G protein-coupled receptors (GPCRs) and Rho GTPases. PKD1 activation in cardiomyocytes occurs through PKC-dependent and -independent mechanisms. In vivo, cardiac PKD1 is activated in multiple rodent models of pathological cardiac remodeling. PKD1 activation correlates with phosphorylation-dependent nuclear export of HDAC5, and reduction of endogenous PKD1 expression with small interfering RNA suppresses HDAC5 shuttling and associated cardiomyocyte growth. Conversely, ectopic overexpression of constitutively active PKD1 in mouse heart leads to dilated cardiomyopathy. These findings support a role for PKD1 in the control of pathological remodeling of the heart via its ability to phosphorylate and neutralize HDAC5.


Subject(s)
Gene Expression Regulation , Myocytes, Cardiac/metabolism , Protein Kinases/physiology , Signal Transduction , Stress, Physiological/metabolism , Animals , Animals, Newborn , COS Cells , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Catalytic Domain , Cells, Cultured , Chlorocebus aethiops , Enzyme Activation , Heart Ventricles/cytology , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Transgenic , Models, Biological , Myocytes, Cardiac/pathology , Protein Kinase C , RNA, Small Interfering/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WF , Rats, Sprague-Dawley
20.
Mol Cell Biol ; 24(24): 10636-49, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15572669

ABSTRACT

Diverse pathological insults trigger a cardiac remodeling process during which myocytes undergo hypertrophy, with consequent decline in cardiac function and eventual heart failure. Multiple transcriptional regulators of pathological cardiac hypertrophy are controlled at the level of subcellular distribution. For example, prohypertrophic transcription factors belonging to the nuclear factor of activated T cells (NFAT) and GATA families are subject to CRM1-dependent nuclear export but are rapidly relocalized to the nucleus in response to cues for hypertrophic growth. Here, we demonstrate that the antihypertrophic chromatin-modifying enzyme histone deacetylase 5 (HDAC5) is shuttled out of the cardiomyocyte nucleus via a CRM1-mediated pathway in response to diverse signals for hypertrophy. CRM1 antagonists block the agonist-mediated nuclear export of HDAC 5 and repress pathological gene expression and associated hypertrophy of cultured cardiomyocytes. Conversely, CRM1 activity is dispensable for nonpathological cardiac gene activation mediated by thyroid hormone and insulin-like growth factor 1, agonists that fail to trigger the nuclear export of HDAC5. These results suggest a selective role for CRM1 in derepression of pathological cardiac genes via its neutralizing effects on antihypertrophic factors such as HDAC5. Pharmacological approaches targeting CRM1-dependent nuclear export in heart muscle may have salutary effects on cardiac function by suppressing maladaptive changes in gene expression evoked by stress signals.


Subject(s)
Cardiomegaly/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation , Karyopherins/metabolism , Myocytes, Cardiac/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Adenoviridae/genetics , Adenylate Kinase/analysis , Adenylate Kinase/metabolism , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/pharmacology , Animals , Animals, Newborn , Antibodies, Monoclonal/metabolism , Atrial Natriuretic Factor/analysis , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/physiology , Cardiomegaly/genetics , Cell Size , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fluoresceins , Fluorescent Dyes , Green Fluorescent Proteins/metabolism , Heart Ventricles/cytology , Histone Deacetylases/metabolism , Immunoblotting , Karyopherins/antagonists & inhibitors , Karyopherins/pharmacology , Microscopy, Fluorescence , Myocytes, Cardiac/cytology , Precipitin Tests , RNA/analysis , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Transcriptional Activation , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...