Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
J Diabetes Res ; 2021: 9979234, 2021.
Article in English | MEDLINE | ID: mdl-34368369

ABSTRACT

OBJECTIVE: To investigate if PP2A plays a role in metformin-induced insulin sensitivity improvement in human skeletal muscle cells. Participants. Eight lean insulin-sensitive nondiabetic participants (4 females and 4 males; age: 21.0 ± 1.0 years; BMI: 22.0 ± 0.7 kg/m2; 2-hour OGTT: 97.0 ± 6.0 mg/dl; HbA1c: 5.3 ± 0.1%; fasting plasma glucose: 87.0 ± 2.0 mg/dl; M value; 11.0 ± 1.0 mg/kgBW/min). DESIGN: A hyperinsulinemic-euglycemic clamp was performed to assess insulin sensitivity in human subjects, and skeletal muscle biopsy samples were obtained. Primary human skeletal muscle cells (shown to retain metabolic characteristics of donors) were cultured from these muscle biopsies that included 8 lean insulin-sensitive participants. Cultured cells were expanded, differentiated into myotubes, and treated with 50 µM metformin for 24 hours before harvesting. PP2Ac activity was measured by a phosphatase activity assay kit (Millipore) according to the manufacturer's protocol. RESULTS: The results indicated that metformin significantly increased the activity of PP2A in the myotubes for all 8 lean insulin-sensitive nondiabetic participants, and the average fold increase is 1.54 ± 0.11 (P < 0.001). CONCLUSIONS: These results provided the first evidence that metformin can activate PP2A in human skeletal muscle cells derived from lean healthy insulin-sensitive participants and may help to understand metformin's action in skeletal muscle in humans.


Subject(s)
Metformin/pharmacology , Muscle Cells/drug effects , Muscle, Skeletal/drug effects , Protein Phosphatase 2/metabolism , Adult , Cells, Cultured , Female , Humans , Insulin Resistance , Male , Muscle Cells/enzymology , Muscle, Skeletal/enzymology , TOR Serine-Threonine Kinases/physiology , Thinness , Young Adult
2.
Cell Mol Immunol ; 18(6): 1463-1475, 2021 06.
Article in English | MEDLINE | ID: mdl-31797905

ABSTRACT

His-tRNA synthetase (HARS) is targeted by autoantibodies in chronic and acute inflammatory anti-Jo-1-positive antisynthetase syndrome. The extensive activation and migration of immune cells into lung and muscle are associated with interstitial lung disease, myositis, and morbidity. It is unknown whether the sequestration of HARS is an epiphenomenon or plays a causal role in the disease. Here, we show that HARS circulates in healthy individuals, but it is largely undetectable in the serum of anti-Jo-1-positive antisynthetase syndrome patients. In cultured primary human skeletal muscle myoblasts (HSkMC), HARS is released in increasing amounts during their differentiation into myotubes. We further show that HARS regulates immune cell engagement and inhibits CD4+ and CD8+ T-cell activation. In mouse and rodent models of acute inflammatory diseases, HARS administration downregulates immune activation. In contrast, neutralization of extracellular HARS by high-titer antibody responses during tissue injury increases susceptibility to immune attack, similar to what is seen in humans with anti-Jo-1-positive disease. Collectively, these data suggest that extracellular HARS is homeostatic in normal subjects, and its sequestration contributes to the morbidity of the anti-Jo-1-positive antisynthetase syndrome.


Subject(s)
Histidine-tRNA Ligase/blood , Immunity , Organ Specificity , Animals , Autoantibodies/blood , Case-Control Studies , Cell Differentiation/drug effects , Disease Models, Animal , Female , Histidine-tRNA Ligase/immunology , Humans , Immunity/drug effects , Immunomodulation/drug effects , Insulin-Like Growth Factor I/pharmacology , Lung/drug effects , Lung/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , Middle Aged , Muscle Cells/drug effects , Muscle Cells/enzymology , Muscles/drug effects , Muscles/pathology , Myositis/blood , Myositis/diagnostic imaging , Myositis/immunology , Organ Specificity/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tomography, X-Ray Computed
3.
Bull Exp Biol Med ; 169(6): 811-814, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33123916

ABSTRACT

The distribution of NO and H2S in the arterial vessels of the anterior abdominal wall after implantation of a polypropylene mesh was studied by immunohistochemical methods at different stages of healing of the surgical wound in mature male Wistar rats. The presence of enzymes of NO and H2S synthesis in the wall of arterial vessels of the soft tissues of the anterior abdominal wall has been established. It has been shown that endothelial NO synthase is localized exclusively in the endothelium of both large and small vessels. Cystathionine γ lyase in small vessels is located only in the endothelial lining, whereas in large arteries and vessels of medium caliber, it is located in the endothelium and in myocytes. Inducible NO synthase appears in the artery wall only in animals with implanted polypropylene mesh by day 5 of the postoperative period, reaching the maximum by day 10. The content and localization of cystathionine γ lyase in the vascular wall of sham-operated and experimental rats did not much differ from the control values.


Subject(s)
Cystathionine gamma-Lyase/genetics , Endothelium, Vascular/drug effects , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type II/genetics , Polypropylenes/pharmacology , Surgical Mesh , Abdominal Wall/blood supply , Abdominal Wall/surgery , Animals , Blood Vessels/cytology , Blood Vessels/drug effects , Blood Vessels/enzymology , Cystathionine gamma-Lyase/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Gene Expression , Hydrogen Sulfide/metabolism , Implants, Experimental , Male , Muscle Cells/cytology , Muscle Cells/drug effects , Muscle Cells/enzymology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Wistar , Wound Healing
4.
FASEB J ; 33(1): 1248-1261, 2019 01.
Article in English | MEDLINE | ID: mdl-30138037

ABSTRACT

Protein glycosylation is an essential posttranslational modification that affects a myriad of physiologic processes. Humans with genetic defects in glycosylation, which result in truncated glycans, often present with significant cardiac deficits. Acquired heart diseases and their associated risk factors were also linked to aberrant glycosylation, highlighting its importance in human cardiac disease. In both cases, the link between causation and corollary remains enigmatic. The glycosyltransferase gene, mannosyl (α-1,3-)-glycoprotein ß-1,2- N-acetylglucosaminyltransferase (Mgat1), whose product, N-acetylglucosaminyltransferase 1 (GlcNAcT1) is necessary for the formation of hybrid and complex N-glycan structures in the medial Golgi, was shown to be at reduced levels in human end-stage cardiomyopathy, thus making Mgat1 an attractive target for investigating the role of hybrid/complex N-glycosylation in cardiac pathogenesis. Here, we created a cardiomyocyte-specific Mgat1 knockout (KO) mouse to establish a model useful in exploring the relationship between hybrid/complex N-glycosylation and cardiac function and disease. Biochemical and glycomic analyses showed that Mgat1KO cardiomyocytes produce predominately truncated N-glycan structures. All Mgat1KO mice died significantly younger than control mice and demonstrated chamber dilation and systolic dysfunction resembling human dilated cardiomyopathy (DCM). Data also indicate that a cardiomyocyte L-type voltage-gated Ca2+ channel (Cav) subunit (α2δ1) is a GlcNAcT1 target, and Mgat1KO Cav activity is shifted to more-depolarized membrane potentials. Consistently, Mgat1KO cardiomyocyte Ca2+ handling is altered and contraction is dyssynchronous compared with controls. The data demonstrate that reduced hybrid/complex N-glycosylation contributes to aberrant cardiac function at whole-heart and myocyte levels drawing a direct link between altered glycosylation and heart disease. Thus, the Mgat1KO provides a model for investigating the relationship between systemic reductions in glycosylation and cardiac disease, showing that clinically relevant changes in cardiomyocyte hybrid/complex N-glycosylation are sufficient to cause DCM and early death.-Ednie, A. R., Deng, W., Yip, K.-P., Bennett, E. S. Reduced myocyte complex N-glycosylation causes dilated cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Muscle Cells/metabolism , Animals , Calcium Channels, L-Type/metabolism , Glycosylation , Ion Channel Gating , Mice , Mice, Knockout , Muscle Cells/enzymology , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism
5.
Biotechnol Lett ; 41(1): 59-68, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30311029

ABSTRACT

OBJECTIVE: Mitochondrial dysfunction plays a pivotal role in various pathophysiological processes of heart. Cytochrome oxidase subunit 6B1 (COX6B1) is a subunit of cytochrome oxidase. METHODS: Cardiomyocytes were isolated from neonatal SD rats (within 24 h of birth) by repeating digestion of collagenase and trypsin. COX6B1 over-expression and hypoxia/reoxygenation was conducted on neonatal rat cardiomyocytes. Cell viability, apoptosis rates, mitochondria membrane potential and mitochondrial permeabilization transition pores (mPTPs) were then determined respectively by Cell performing Counting Kit-8 (CCK-8), Annexin-V/PI assay, JC-1 assay, mPTP assay. The expression of cyto C and apoptosis-related factors were detected by RT-Qpcr and Western blot. RESULTS: Hypoxia/reoxygenation increased apoptosis and mPTP levels, and decreased mitochondria membrane potential in I/R and I/R + EV groups. COX6B1 over-expression increased mitochondria cyto C, pro-caspase-3, pro-caspase-9 and bcl-2, while it decreased cytosol cyto C, cleaved-caspase-3, cleaved-caspase-9 and bax compared to I/R + EV group. CONCLUSION: COX6B1 protected cardiomyocytes from hypoxia/reoxygenation injury by reducing ROS production and cell apoptosis, during which reduction of the release of cytochrome C from mitochondria to cytosol was involved. Our study demonstrated that COX6B1 may be an candidate target gene in preventing hypoxia/reoxygenation injury of cardiomyocytes.


Subject(s)
Electron Transport Complex IV/biosynthesis , Gene Expression Regulation, Enzymologic , Mitochondria, Heart/enzymology , Muscle Cells/enzymology , Myocardial Reperfusion Injury/enzymology , Animals , Animals, Newborn , Female , Mitochondria, Heart/pathology , Muscle Cells/pathology , Myocardial Reperfusion Injury/pathology , Rats , Rats, Sprague-Dawley
6.
J Neuropathol Exp Neurol ; 78(2): 191-194, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30590671

ABSTRACT

Cerebral small vessel disease in deep penetrating arteries is a major cause of lacunar infarcts, white matter lesions and vascular cognitive impairment. Local cerebral blood flow in these small vessels is controlled by endothelial-derived nitric oxide, which exerts a primary vasodilator stimulus on vascular myocytes, via cytoplasmic cyclic GMP. Here, we investigated whether the cGMP-degrading enzyme phosphodiesterase-5 (PDE5) is present in small penetrating arteries in the deep subcortical white matter of older people. Frontal cortical tissue blocks were examined from donated brains of older people (n = 42, 24 male: 18 female, median age 81, range: 59-100 years). PDE5, detected by immunohistochemical labeling, was graded as absent, sparse, or abundant in vascular cells within small arteries in subcortical white matter (vessel outer diameter: 20-100 µm). PDE5 labeling within arterial myocytes was detected in all cases. Degree of PDE5 expression (absent, sparse, or abundant) was not associated with age or with neuropathological diagnosis of small vessel disease. In conclusion, PDE5 is present in vascular myocytes within small penetrating arteries in older people. This is a potential molecular target for pharmacological interventions.


Subject(s)
Brain/enzymology , Cerebral Arteries/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Muscle, Smooth, Vascular/enzymology , White Matter/enzymology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Muscle Cells/enzymology
7.
J Neurosci ; 38(35): 7683-7700, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30054395

ABSTRACT

Aging contributes to cellular stress and neurodegeneration. Our understanding is limited regarding the tissue-restricted mechanisms providing protection in postmitotic cells throughout life. Here, we show that spinal cord motoneurons exhibit a high abundance of asymmetric dimethyl arginines (ADMAs) and the presence of this posttranslational modification provides protection against environmental stress. We identify protein arginine methyltransferase 8 (PRMT8) as a tissue-restricted enzyme responsible for proper ADMA level in postmitotic neurons. Male PRMT8 knock-out mice display decreased muscle strength with aging due to premature destabilization of neuromuscular junctions. Mechanistically, inhibition of methyltransferase activity or loss of PRMT8 results in accumulation of unrepaired DNA double-stranded breaks and decrease in the cAMP response-element-binding protein 1 (CREB1) level. As a consequence, the expression of CREB1-mediated prosurvival and regeneration-associated immediate early genes is dysregulated in aging PRMT8 knock-out mice. The uncovered role of PRMT8 represents a novel mechanism of stress tolerance in long-lived postmitotic neurons and identifies PRMT8 as a tissue-specific therapeutic target in the prevention of motoneuron degeneration.SIGNIFICANCE STATEMENT Although most of the cells in our body have a very short lifespan, postmitotic neurons must survive for many decades. Longevity of a cell within the organism depends on its ability to properly regulate signaling pathways that counteract perturbations, such as DNA damage, oxidative stress, or protein misfolding. Here, we provide evidence that tissue-specific regulators of stress tolerance exist in postmitotic neurons. Specifically, we identify protein arginine methyltransferase 8 (PRMT8) as a cell-type-restricted arginine methyltransferase in spinal cord motoneurons (MNs). PRMT8-dependent arginine methylation is required for neuroprotection against age-related increased of cellular stress. Tissue-restricted expression and the enzymatic activity of PRMT8 make it an attractive target for drug development to delay the onset of neurodegenerative disorders.


Subject(s)
DNA Damage/physiology , Motor Neurons/enzymology , Protein-Arginine N-Methyltransferases/physiology , Aging/metabolism , Amino Acid Sequence , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/physiology , DNA Breaks, Double-Stranded , DNA Repair , Isometric Contraction , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle Cells/enzymology , Muscle Cells/physiology , Neuromuscular Junction/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/deficiency , Protein-Arginine N-Methyltransferases/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/metabolism , Reflex, Abnormal , Rotarod Performance Test , Spinal Cord/cytology , Spinal Cord/growth & development
8.
PLoS One ; 13(5): e0197254, 2018.
Article in English | MEDLINE | ID: mdl-29758057

ABSTRACT

Ankyrin repeat and kinase domain containing 1 (ANKK1) gene has been widely related to neuropsychiatry disorders. The localization of ANKK1 in neural progenitors and its correlation with the cell cycle has suggested its participation in development. However, ANKK1 functions still need to be identified. Here, we have further characterized the ANKK1 localization in vivo and in vitro, by using immunolabeling, quantitative real-time PCR and Western blot in the myogenic lineage. Histologic investigations in mice and humans revealed that ANKK1 is expressed in precursors of embryonic and adult muscles. In mice embryos, ANKK1 was found in migrating myotubes where it shows a polarized cytoplasmic distribution, while proliferative myoblasts and satellite cells show different isoforms in their nuclei and cytoplasm. In vitro studies of ANKK1 protein isoforms along the myogenic progression showed the decline of nuclear ANKK1-kinase until its total exclusion in myotubes. In adult mice, ANKK1 was expressed exclusively in the Fast-Twitch muscles fibers subtype. The induction of glycolytic metabolism in C2C12 cells with high glucose concentration or treatment with berberine caused a significant increase in the ANKK1 mRNA. Similarly, C2C12 cells under hypoxic conditions caused the increase of nuclear ANKK1. These results altogether show a relationship between ANKK1 gene regulation and the metabolism of muscles during development and in adulthood. Finally, we found ANKK1 expression in regenerative fibers of muscles from dystrophic patients. Future studies in ANKK1 biology and the pathological response of muscles will reveal whether this protein is a novel muscle disease biomarker.


Subject(s)
Muscle Cells/enzymology , Muscle, Skeletal/enzymology , Protein Serine-Threonine Kinases/metabolism , Stem Cells/enzymology , Adult , Animals , Cell Hypoxia/physiology , Cell Line , Cell Nucleus/enzymology , Cell Proliferation , Child , Cytoplasm/enzymology , Female , Humans , Infant , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Muscle Cells/cytology , Muscle Cells/pathology , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Muscular Dystrophies/enzymology , Muscular Dystrophies/pathology , Stem Cells/cytology , Stem Cells/pathology
9.
Subcell Biochem ; 87: 229-258, 2018.
Article in English | MEDLINE | ID: mdl-29464562

ABSTRACT

The calcium pump (a.k.a. Ca2+-ATPase or SERCA) is a membrane transport protein ubiquitously found in the endoplasmic reticulum (ER) of all eukaryotic cells. As a calcium transporter, SERCA maintains the low cytosolic calcium level that enables a vast array of signaling pathways and physiological processes (e.g. synaptic transmission, muscle contraction, fertilization). In muscle cells, SERCA promotes relaxation by pumping calcium ions from the cytosol into the lumen of the sarcoplasmic reticulum (SR), the main storage compartment for intracellular calcium. X-ray crystallographic studies have provided an extensive understanding of the intermediate states that SERCA populates as it progresses through the calcium transport cycle. Historically, SERCA is also known to be regulated by small transmembrane peptides, phospholamban (PLN) and sarcolipin (SLN). PLN is expressed in cardiac muscle, whereas SLN predominates in skeletal and atrial muscle. These two regulatory subunits play critical roles in cardiac contractility. While our understanding of these regulatory mechanisms are still developing, SERCA and PLN are one of the best understood examples of peptide-transporter regulatory interactions. Nonetheless, SERCA appeared to have only two regulatory subunits, while the related sodium pump (a.k.a. Na+, K+-ATPase) has at least nine small transmembrane peptides that provide tissue specific regulation. The last few years have seen a renaissance in our understanding of SERCA regulatory subunits. First, structures of the SERCA-SLN and SERCA-PLN complexes revealed molecular details of their interactions. Second, an array of micropeptides concealed within long non-coding RNAs have been identified as new SERCA regulators. This chapter will describe our current understanding of SERCA structure, function, and regulation.


Subject(s)
Calcium Signaling/physiology , Calcium , Endoplasmic Reticulum , Muscle Cells/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animals , Calcium/chemistry , Calcium/metabolism , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Crystallography, X-Ray , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Humans , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Proteolipids/chemistry , Proteolipids/genetics , Proteolipids/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
10.
J Cell Physiol ; 233(12): 9345-9353, 2018 12.
Article in English | MEDLINE | ID: mdl-29319158

ABSTRACT

This study was conceived to evaluate the effects of three different diets on body composition, metabolic parameters, and serum oxidative status. We enrolled three groups of healthy men (omnivores, vegetarians, and vegans) with similar age, weight and BMI, and we observed a significant decrease in muscle mass index and lean body mass in vegan compared to vegetarian and omnivore groups, and higher serum homocysteine levels in vegetarians and vegans compared to omnivores. We studied whether serum from omnivore, vegetarian, and vegan subjects affected oxidative stress, growth and differentiation of both cardiomyoblast cell line H9c2 and H-H9c2 (H9c2 treated with H2 O2 to induce oxidative damage). We demonstrated that vegan sera treatment of both H9c2 and H-H9c2 cells induced an increase of TBARS values and cell death and a decrease of free NO2- compared to vegetarian and omnivorous sera. Afterwards, we investigated the protective effects of vegan, vegetarian, and omnivore sera on the morphological changes induced by H2 O2 in H9c2 cell line. We showed that the omnivorous sera had major antioxidant and differentiation properties compared to vegetarian and vegan sera. Finally, we evaluated the influence of the three different groups of sera on MAPKs pathway and our data suggested that ERK expression increased in H-H9c2 cells treated with vegetarian and vegan sera and could promote cell death. The results obtained in this study demonstrated that restrictive vegan diet could not prevent the onset of metabolic and cardiovascular diseases nor protect by oxidative damage.


Subject(s)
Cell Differentiation , Diet, Vegan , Muscle Cells/cytology , Muscles/anatomy & histology , Adult , Animals , Anthropometry , Cell Count , Cell Line , Cell Shape , Humans , MAP Kinase Signaling System , Male , Muscle Cells/enzymology , Myocytes, Cardiac/pathology , Organ Size , Oxidation-Reduction , Oxidative Stress , Pilot Projects , Rats , Vegetarians , Young Adult
11.
ACS Infect Dis ; 3(8): 585-594, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28605587

ABSTRACT

MDL-860 is a broad-spectrum antipicornavirus compound discovered in 1982 and one of the few promising candidates effective in in vivo virus infection. Despite the effectiveness, the target and the mechanism of action of MDL-860 remain unknown. Here, we have characterized antipoliovirus activity of MDL-860 and identified host phosphatidylinositol-4 kinase III beta (PI4KB) as the target. MDL-860 treatment caused covalent modification and irreversible inactivation of PI4KB. A cysteine residue at amino acid 646 of PI4KB, which locates at the bottom of a surface pocket apart from the active site, was identified as the target site of MDL-860. This work reveals the mechanism of action of this class of PI4KB inhibitors and offers insights into novel allosteric regulation of PI4KB activity.


Subject(s)
Antiviral Agents/pharmacology , Host-Pathogen Interactions/drug effects , Muscle Cells/drug effects , Nitriles/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Allosteric Regulation , Allosteric Site , Antiviral Agents/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression , HEK293 Cells , Humans , Kinetics , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Models, Molecular , Muscle Cells/enzymology , Muscle Cells/virology , Nitriles/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Picornaviridae/drug effects , Picornaviridae/physiology , Protein Binding , Virus Replication/drug effects
12.
Biophys J ; 111(6): 1192-1202, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27653478

ABSTRACT

The cardiac sarco/endoplasmic reticulum calcium ATPase (SERCA) establishes the intracellular calcium gradient across the sarcoplasmic reticulum membrane. It has been proposed that SERCA forms homooligomers that increase the catalytic rate of calcium transport. We investigated SERCA dimerization in rabbit left ventricular myocytes using a photoactivatable cross-linker. Western blotting of cross-linked SERCA revealed higher-molecular-weight species consistent with SERCA oligomerization. Fluorescence resonance energy transfer measurements in cells transiently transfected with fluorescently labeled SERCA2a revealed that SERCA readily forms homodimers. These dimers formed in the absence or presence of the SERCA regulatory partner, phospholamban (PLB) and were unaltered by PLB phosphorylation or changes in calcium or ATP. Fluorescence lifetime data are compatible with a model in which PLB interacts with a SERCA homodimer in a stoichiometry of 1:2. Together, these results suggest that SERCA forms constitutive homodimers in live cells and that dimer formation is not modulated by SERCA conformational poise, PLB binding, or PLB phosphorylation.


Subject(s)
Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Calcium/chemistry , Calcium/metabolism , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Dogs , Fluorescence Resonance Energy Transfer , HEK293 Cells , Heart Ventricles/chemistry , Heart Ventricles/enzymology , Humans , Immunoprecipitation , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Models, Molecular , Muscle Cells/chemistry , Muscle Cells/enzymology , Mutation , Phosphorylation , Photobleaching , Protein Multimerization , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
13.
Biochim Biophys Acta ; 1861(2): 119-129, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26603903

ABSTRACT

Phosphatidylethanolamine N-methyltransferase (PEMT) converts phosphatidylethanolamine (PE) to phosphatidylcholine (PC) in the liver. Mice lacking PEMT are protected from high-fat diet-induced obesity and insulin resistance, and exhibit increased whole-body energy expenditure and oxygen consumption. Since skeletal muscle is a major site of fatty acid oxidation and energy utilization, we determined if rates of fatty acid oxidation/oxygen consumption in muscle are higher in Pemt(-/-) mice than in Pemt(+/+) mice. Although PEMT is abundant in the liver, PEMT protein and activity were undetectable in four types of skeletal muscle. Moreover, amounts of PC and PE in the skeletal muscle were not altered by PEMT deficiency. Thus, we concluded that any influence of PEMT deficiency on skeletal muscle would be an indirect consequence of lack of PEMT in liver. Neither the in vivo rate of fatty acid uptake by muscle nor the rate of fatty acid oxidation in muscle explants and cultured myocytes depended upon Pemt genotype. Nor did PEMT deficiency increase oxygen consumption or respiratory function in skeletal muscle mitochondria. Thus, the increased whole body oxygen consumption in Pemt(-/-) mice, and resistance of these mice to diet-induced weight gain, are not primarily due to increased capacity of skeletal muscle for utilization of fatty acids as an energy source.


Subject(s)
Fatty Acids/metabolism , Liver/enzymology , Muscle, Skeletal/enzymology , Obesity/enzymology , Phosphatidylcholines/metabolism , Phosphatidylethanolamine N-Methyltransferase/deficiency , Phosphatidylethanolamines/metabolism , Animals , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Energy Metabolism , Gene Expression , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/enzymology , Muscle Cells/cytology , Muscle Cells/enzymology , Obesity/etiology , Obesity/genetics , Oxidation-Reduction , Oxygen Consumption , Phosphatidylethanolamine N-Methyltransferase/genetics , Primary Cell Culture
14.
Food Funct ; 6(9): 3056-64, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26205038

ABSTRACT

Muscle wasting is associated with a variety of chronic or inflammatory disorders. Evidence suggests that inflammatory cytokines play a vital role in muscle inflammatory pathology and this may result in oxidative damage and mitochondrial dysfunction in skeletal muscle. In our study, we used microwave degradation to prepare a water-soluble low molecular weight guluronate (LMG) of 3000 Da from Fucus vesiculosus obtained from Canada, the Atlantic Ocean. We demonstrated the structural characteristics, using HPLC, FTIR and NMR of LMG and investigated its effects on oxidative damage and mitochondrial dysfunction in C2C12 skeletal muscle cells induced by tumor necrosis factor alpha (TNF-α), a cell inflammatory cytokine. The results indicated that LMG could alleviate mitochondrial reactive oxygen species (ROS) production, increase the activities of antioxidant enzymes (GSH and SOD), promote mitochondrial membrane potential (MMP) and upregulate the expression of mitochondrial respiratory chain protein in TNF-α-induced C2C12 cells. LMG supplement also increased the mitochondrial DNA copy number and mitochondrial biogenesis related genes in TNF-α-induced C2C12 cells. LMG may exert these protective effects through the nuclear factor kappa B (NF-κB) signaling pathway. These suggest that LMG is capable of protecting TNF-α-induced C2C12 cells against oxidative damage and mitochondrial dysfunction.


Subject(s)
Cachexia/metabolism , Fucus/chemistry , Hexuronic Acids/pharmacology , Mitochondria/drug effects , Muscle Cells/drug effects , Muscle, Skeletal/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Cachexia/drug therapy , Cachexia/enzymology , Cell Line , Glutathione/metabolism , Hexuronic Acids/chemistry , Humans , Mice , Mitochondria/metabolism , Molecular Weight , Muscle Cells/enzymology , Muscle Cells/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Reactive Oxygen Species/metabolism , Seaweed/chemistry , Superoxide Dismutase/metabolism
15.
Nature ; 519(7544): 472-6, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25799991

ABSTRACT

Cyclic guanosine monophosphate (cGMP) is a second messenger molecule that transduces nitric-oxide- and natriuretic-peptide-coupled signalling, stimulating phosphorylation changes by protein kinase G. Enhancing cGMP synthesis or blocking its degradation by phosphodiesterase type 5A (PDE5A) protects against cardiovascular disease. However, cGMP stimulation alone is limited by counter-adaptions including PDE upregulation. Furthermore, although PDE5A regulates nitric-oxide-generated cGMP, nitric oxide signalling is often depressed by heart disease. PDEs controlling natriuretic-peptide-coupled cGMP remain uncertain. Here we show that cGMP-selective PDE9A (refs 7, 8) is expressed in the mammalian heart, including humans, and is upregulated by hypertrophy and cardiac failure. PDE9A regulates natriuretic-peptide- rather than nitric-oxide-stimulated cGMP in heart myocytes and muscle, and its genetic or selective pharmacological inhibition protects against pathological responses to neurohormones, and sustained pressure-overload stress. PDE9A inhibition reverses pre-established heart disease independent of nitric oxide synthase (NOS) activity, whereas PDE5A inhibition requires active NOS. Transcription factor activation and phosphoproteome analyses of myocytes with each PDE selectively inhibited reveals substantial differential targeting, with phosphorylation changes from PDE5A inhibition being more sensitive to NOS activation. Thus, unlike PDE5A, PDE9A can regulate cGMP signalling independent of the nitric oxide pathway, and its role in stress-induced heart disease suggests potential as a therapeutic target.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cardiomegaly/enzymology , Cardiomegaly/metabolism , Cyclic GMP/metabolism , Nitric Oxide , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/deficiency , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Aortic Valve Stenosis/complications , Cardiomegaly/drug therapy , Cardiomegaly/etiology , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Cells/enzymology , Myocardium/enzymology , Natriuretic Peptides/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Pressure , Signal Transduction/drug effects , Stress, Physiological , Up-Regulation
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(3): 312-5, 320, 2015 Mar.
Article in Chinese | MEDLINE | ID: mdl-25744833

ABSTRACT

OBJECTIVE: To observe changes of inflammatory mediators, histone deacetylase 2 (HDAC2) and nuclear factor κB (NF-κB) in murine C2C12 skeletal muscle myocytes after exposed to cigarette smoke. METHODS: Murine C2C12 skeletal muscle myocytes were cultured and treated with cigarette smoke extract (CSE). MTT assay was used to detect the effect of CSE on cell proliferation to determine appropriate concentration of CSE. The C2C12 cells cultured for 6-7 days were planted in six-well plates, and divided into control group, (6.25, 12.50, 25.0) mL/L CSE groups. The cells were cultured for 24 hours. The levels of interleukin-8 (IL-8) and tumor necrosis factor-α (TNF-α) in the supernatant were measured by ELISA. The mRNA level of HDAC2 was determined by real-time quantitative PCR. The protein level of HDAC2 was detected by Western blotting. HDAC2/NF-κB compound was determined by the method of co-immunoprecipitation. RESULTS: MTT assay showed that CSE at the concentration of 50 mL/L inhibited proliferation of C2C12 cells. After 24-hour treatment with CSE, IL-8 and TNF-α releasing from C2C12 cells increased and the level of HDAC2 mRNA and protein were reduced, which were CSE dose-dependent. Co-immunoprecipitation confirmed that HDAC2/NF-κB compound existed in the CSE-exposed C2C12 cells. CONCLUSION: CSE can down-regulate the expression of HDAC2 and increase inflammatory cytokines releasing from C2C12 cells.


Subject(s)
Histone Deacetylase 2/genetics , Interleukin-8/metabolism , Muscle Cells/drug effects , Muscle, Skeletal/drug effects , NF-kappa B/metabolism , Nicotiana/chemistry , Smoke/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Proliferation/drug effects , Down-Regulation/drug effects , Histone Deacetylase 2/metabolism , Interleukin-8/genetics , Mice , Muscle Cells/cytology , Muscle Cells/enzymology , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , NF-kappa B/genetics , Tumor Necrosis Factor-alpha/genetics
17.
J Agric Food Chem ; 62(19): 4298-305, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24712520

ABSTRACT

The mitochondrion is a central organelle in cellular energy homeostasis; thus, reduced mitochondrial activity has been associated with aging and metabolic disorders. This paper provides biological evidence that coumestrol, which is a natural isoflavone, activates mitochondrial biogenesis. In cultured myocytes, coumestrol activated the silent information regulator two ortholog 1 (Sirt1) through the elevation of the intracellular NAD(+)/NADH ratio. Coumestrol also increased the mitochondrial contents and induced the expression of key proteins in the mitochondrial electron transfer chain in cultured myocytes. A Sirt1 inhibitor and Sirt1-targeting siRNAs abolished the effect of coumestrol on mitochondrial biogenesis. Similar to an increase in mitochondrial content, coumestrol improved myocyte function with increased ATP concentration. Taken together, the data suggest that coumestrol is a novel inducer of mitochondrial biogenesis through the activation of Sirt1.


Subject(s)
Coumestrol/pharmacology , Mitochondria/enzymology , Muscle Cells/drug effects , Muscle, Skeletal/cytology , Sirtuin 1/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Mice , Mitochondria/drug effects , Muscle Cells/enzymology , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Sirtuin 1/genetics
18.
Mol Biol Cell ; 25(7): 1159-70, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478457

ABSTRACT

Rab-GTPases are important molecular switches regulating intracellular vesicle traffic, and we recently showed that Rab8A and Rab13 are activated by insulin in muscle to mobilize GLUT4-containing vesicles to the muscle cell surface. Here we show that the unconventional motor protein myosin Va (MyoVa) is an effector of Rab8A in this process. In CHO-IR cell lysates, a glutathione S-transferase chimera of the cargo-binding COOH tail (CT) of MyoVa binds Rab8A and the related Rab10, but not Rab13. Binding to Rab8A is stimulated by insulin in a phosphatidylinositol 3-kinase-dependent manner, whereas Rab10 binding is insulin insensitive. MyoVa-CT preferentially binds GTP-locked Rab8A. Full-length green fluorescent protein (GFP)-MyoVa colocalizes with mCherry-Rab8A in perinuclear small puncta, whereas GFP-MyoVa-CT collapses the GTPase into enlarged perinuclear depots. Further, GFP-MyoVa-CT blocks insulin-stimulated translocation of exofacially myc-tagged GLUT4 to the surface of muscle cells. Mutation of amino acids in MyoVa-CT predicted to bind Rab8A abrogates both interaction with Rab8A (not Rab10) and inhibition of insulin-stimulated GLUT4myc translocation. Of importance, small interfering RNA-mediated MyoVa silencing reduces insulin-stimulated GLUT4myc translocation. Rab8A colocalizes with GLUT4 in perinuclear but not submembrane regions visualized by confocal total internal reflection fluorescence microscopy. Hence insulin signaling to the molecular switch Rab8A connects with the motor protein MyoVa to mobilize GLUT4 vesicles toward the muscle cell plasma membrane.


Subject(s)
Exocytosis/drug effects , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Muscle Cells/metabolism , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Secretory Vesicles/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Binding Sites , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Knockdown Techniques , Gene Silencing/drug effects , Guanosine Triphosphate/pharmacology , Humans , Mice , Muscle Cells/drug effects , Muscle Cells/enzymology , Mutation/genetics , Myoblasts/drug effects , Myoblasts/metabolism , Myosin Heavy Chains/chemistry , Myosin Type V/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Secretory Vesicles/drug effects
19.
Food Chem ; 147: 124-30, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24206695

ABSTRACT

Soy peptides consumption has been associated with beneficial effects in type 2 diabetes patients. However, the peptide fractions responsible for these effects, and their mechanisms of action, have not been identified yet. In this study, we have isolated soybean peptides by electrodialysis with an ultrafiltration membrane (EDUF) at 50 V/100 kDa, and tested them for their capacity to improve glucose uptake in L6 muscle cells. We observed that these fractions were able to significantly enhance glucose uptake in the presence of insulin. The reported bioactivity would be due to the low molecular weight peptides (300-500 Da) recovered. Moreover, we observed that an enhancement of glucose uptake was correlated to the activation of the AMPK enzyme, well known for its capacity to increase glucose uptake in muscle cells. To our knowledge, this is the first time that bioactive peptides with glucose uptake activity have been isolated from a complex soy matrix, and that the implication of AMPK in it is demonstrated.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Glycine max/chemistry , Muscle Cells/metabolism , Peptides/metabolism , Biological Transport , Dialysis , Humans , Molecular Weight , Muscle Cells/enzymology , Peptides/chemistry , Peptides/isolation & purification , Signal Transduction , Ultrafiltration/instrumentation
20.
Elife ; 2: e01503, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24368734

ABSTRACT

Enhancers play a central role in cell-type-specific gene expression and are marked by H3K4me1/2. Active enhancers are further marked by H3K27ac. However, the methyltransferases responsible for H3K4me1/2 on enhancers remain elusive. Furthermore, how these enzymes function on enhancers to regulate cell-type-specific gene expression is unclear. In this study, we identify MLL4 (KMT2D) as a major mammalian H3K4 mono- and di-methyltransferase with partial functional redundancy with MLL3 (KMT2C). Using adipogenesis and myogenesis as model systems, we show that MLL4 exhibits cell-type- and differentiation-stage-specific genomic binding and is predominantly localized on enhancers. MLL4 co-localizes with lineage-determining transcription factors (TFs) on active enhancers during differentiation. Deletion of Mll4 markedly decreases H3K4me1/2, H3K27ac, Mediator and Polymerase II levels on enhancers and leads to severe defects in cell-type-specific gene expression and cell differentiation. Together, these findings identify MLL4 as a major mammalian H3K4 mono- and di-methyltransferase essential for enhancer activation during cell differentiation. DOI: http://dx.doi.org/10.7554/eLife.01503.001.


Subject(s)
Adipocytes/enzymology , Adipogenesis , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Muscle Cells/enzymology , Muscle Development , 3T3-L1 Cells , Animals , Binding Sites , Cell Lineage , Computational Biology , DNA Polymerase II/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Genomics/methods , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Mediator Complex/metabolism , Methylation , Mice , Mice, Knockout , Promoter Regions, Genetic , Time Factors , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...