Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Circ Res ; 133(1): 6-21, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37232152

ABSTRACT

BACKGROUND: Obesity induces cardiomyopathy characterized by hypertrophy and diastolic dysfunction. Whereas mitophagy mediated through an Atg7 (autophagy related 7)-dependent mechanism serves as an essential mechanism to maintain mitochondrial quality during the initial development of obesity cardiomyopathy, Rab9 (Ras-related protein Rab-9A)-dependent alternative mitophagy takes over the role during the chronic phase. Although it has been postulated that DRP1 (dynamin-related protein 1)-mediated mitochondrial fission and consequent separation of the damaged portions of mitochondria are essential for mitophagy, the involvement of DRP1 in mitophagy remains controversial. We investigated whether endogenous DRP1 is essential in mediating the 2 forms of mitophagy during high-fat diet (HFD)-induced obesity cardiomyopathy and, if so, what the underlying mechanisms are. METHODS: Mice were fed either a normal diet or an HFD (60 kcal %fat). Mitophagy was evaluated using cardiac-specific Mito-Keima mice. The role of DRP1 was evaluated using tamoxifen-inducible cardiac-specific Drp1knockout (Drp1 MCM) mice. RESULTS: Mitophagy was increased after 3 weeks of HFD consumption. The induction of mitophagy by HFD consumption was completely abolished in Drp1 MCM mouse hearts, in which both diastolic and systolic dysfunction were exacerbated. The increase in LC3 (microtubule-associated protein 1 light chain 3)-dependent general autophagy and colocalization between LC3 and mitochondrial proteins was abolished in Drp1 MCM mice. Activation of alternative mitophagy was also completely abolished in Drp1 MCM mice during the chronic phase of HFD consumption. DRP1 was phosphorylated at Ser616, localized at the mitochondria-associated membranes, and associated with Rab9 and Fis1 (fission protein 1) only during the chronic, but not acute, phase of HFD consumption. CONCLUSIONS: DRP1 is an essential factor in mitochondrial quality control during obesity cardiomyopathy that controls multiple forms of mitophagy. Although DRP1 regulates conventional mitophagy through a mitochondria-associated membrane-independent mechanism during the acute phase, it acts as a component of the mitophagy machinery at the mitochondria-associated membranes in alternative mitophagy during the chronic phase of HFD consumption.


Subject(s)
Cardiomyopathies , Mitophagy , Animals , Mice , Autophagy/physiology , Cardiomyopathies/genetics , Dynamins/genetics , Dynamins/metabolism , Heart , Mitochondrial Dynamics , Mitophagy/physiology , Obesity/genetics
2.
Am J Physiol Cell Physiol ; 322(2): C260-C274, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34986021

ABSTRACT

Duchenne muscular dystrophy (DMD) is an inherited muscle wasting disease. Metabolic impairments and oxidative stress are major secondary mechanisms that severely worsen muscle function in DMD. Here, we sought to determine whether germline reduction or ablation of sarcolipin (SLN), an inhibitor of sarco/endoplasmic reticulum (SR) Ca2+ ATPase (SERCA), improves muscle metabolism and ameliorates muscle pathology in the mdx mouse model of DMD. Glucose and insulin tolerance tests show that glucose clearance rate and insulin sensitivity were improved in the SLN haploinsufficient mdx (mdx:sln+/-) and SLN-deficient mdx (mdx:sln-/-) mice. The histopathological analysis shows that fibrosis and necrosis were significantly reduced in muscles of mdx:sln+/- and mdx:sln-/- mice. SR Ca2+ uptake, mitochondrial complex protein levels, complex activities, mitochondrial Ca2+ uptake and release, and mitochondrial metabolism were significantly improved, and lipid peroxidation and protein carbonylation were reduced in the muscles of mdx:sln+/- and mdx:sln-/- mice. These data demonstrate that reduction or ablation of SLN expression can improve muscle metabolism, reduce oxidative stress, decrease muscle pathology, and protects the mdx mice from glucose intolerance.


Subject(s)
Muscle Proteins/antagonists & inhibitors , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Proteolipids/antagonists & inhibitors , Proteolipids/biosynthesis , Animals , Blood Glucose/genetics , Blood Glucose/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscle Proteins/genetics , Oxidative Stress/physiology , Proteolipids/genetics
3.
Am J Physiol Heart Circ Physiol ; 320(1): H200-H210, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33216625

ABSTRACT

Sarcolipin (SLN) is an inhibitor of sarco/endoplasmic reticulum (SR) Ca2+-ATPase (SERCA) and expressed at high levels in the ventricles of animal models for and patients with Duchenne muscular dystrophy (DMD). The goal of this study was to determine whether the germline ablation of SLN expression improves cardiac SERCA function and intracellular Ca2+ (Ca2+i) handling and prevents cardiomyopathy in the mdx mouse model of DMD. Wild-type, mdx, SLN-haploinsufficient mdx (mdx:sln+/-), and SLN-deficient mdx (mdx:sln-/-) mice were used for this study. SERCA function and Ca2+i handling were determined by Ca2+ uptake assays and by measuring single-cell Ca2+ transients, respectively. Age-dependent disease progression was determined by histopathological examinations and by echocardiography in 6-, 12-, and 20-mo-old mice. Gene expression changes in the ventricles of mdx:sln+/- mice were determined by RNA-Seq analysis. SERCA function and Ca2+i cycling were improved in the ventricles of mdx:sln+/- mice. Fibrosis and necrosis were significantly decreased, and cardiac function was enhanced in the mdx:sln+/- mice until the study endpoint. The mdx:sln-/- mice also exhibited similar beneficial effects. RNA-Seq analysis identified distinct gene expression changes including the activation of the apelin pathway in the ventricles of mdx:sln+/- mice. Our findings suggest that reducing SLN expression is sufficient to improve cardiac SERCA function and Ca2+i cycling and prevent cardiomyopathy in mdx mice.NEW & NOTEWORTHY First, reducing sarcopolin (SLN) expression improves sarco/endoplasmic reticulum Ca2+ uptake and intracellular Ca2+ handling and prevents cardiomyopathy in mdx mice. Second, reducing SLN expression prevents diastolic dysfunction and improves cardiac contractility in mdx mice Third, reducing SLN expression activates apelin-mediated cardioprotective signaling pathways in mdx heart.


Subject(s)
Cardiomyopathies/prevention & control , Haploinsufficiency , Muscle Proteins/deficiency , Muscular Dystrophy, Duchenne/complications , Myocardium/metabolism , Proteolipids/deficiency , Animals , Apelin/genetics , Apelin/metabolism , Calcium/metabolism , Calcium Signaling , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Disease Models, Animal , Female , Fibrosis , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscle Proteins/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Myocardium/pathology , Necrosis , Proteolipids/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Ventricular Function, Left
4.
Arch Microbiol ; 203(7): 4189-4199, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34076737

ABSTRACT

Bacillus velezensis is widely known for its inherent biosynthetic potential to produce a wide range of bio-macromolecules and secondary metabolites, including polyketides (PKs) and siderophores, as well as ribosomally and non-ribosomally synthesized peptides. In the present study, we aimed to investigate the bio-macromolecules, such as proteins and peptides of Bacillus velezensis strains, namely A6 and P42 by whole-cell sequencing and highlighted the potential application in controlling phytopathogens. The bioactive compounds, specifically secondary metabolites, were characterized by whole-cell protein profiling, Thin-Layer Chromatography, Infra-Red Spectroscopy, Nuclear Magnetic Resonance, Gas Chromatograph and Electro Spray Liquid Chromatography. Gas Chromatography analysis revealed that the A6 and P42 strains exert different functional groups of compounds, such as aromatic ring, aliphatic, alkene, ketone, amine groups and carboxylic acid. Whole-cell protein profiling of A6 and P42 strains of B. velezensis by nano-ESI LC-MS/MS revealed the presence of 945 and 5303 proteins, respectively. The in vitro evaluation of crude extracts (10%) of A6 and P42 significantly inhibited the rice pathogen, Magnaporthe oryzae (MG01), whereas the cell-free culture filtrate (75%) of strain P42 showed 58.97% inhibition. Similarly, in vitro evaluation of crude extract (10%) of P42 strain inhibited bacterial blight of pomegranate pathogen, Xanthomonas axonopodis pv. punicae, which eventually resulted in a higher inhibition zone of 3 cm, whereas the cell-free extract (75%) of the same strain significantly suppressed the growth of the pathogen with an inhibition zone of 1.48 cm. From the results obtained, the crude secondary metabolites and cell-free filtrates (containing bio-macromolecules) of the strains A6 and P42 of B. velezensis can be employed for controlling the bacterial and fungal pathogens of crop plants.


Subject(s)
Ascomycota , Bacillus , Plant Diseases , Xanthomonas axonopodis , Antifungal Agents/chemistry , Antifungal Agents/isolation & purification , Antifungal Agents/pharmacology , Ascomycota/drug effects , Bacillus/chemistry , Chromatography, Liquid , Oryza/microbiology , Pest Control, Biological , Plant Diseases/microbiology , Plant Diseases/prevention & control , Pomegranate/microbiology , Tandem Mass Spectrometry , Xanthomonas axonopodis/drug effects
5.
Mol Ther ; 28(3): 845-854, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31981493

ABSTRACT

Loss of dystrophin leads to Duchenne muscular dystrophy (DMD). A pathogenic feature of DMD is the significant elevation of cytosolic calcium. Supraphysiological calcium triggers protein degradation, membrane damage, and eventually muscle death and dysfunction. Sarcoplasmic/endoplasmic reticulum (SR) calcium ATPase (SERCA) is a calcium pump that transports cytosolic calcium to the SR during excitation-contraction coupling. We hypothesize that a single systemic delivery of SERCA2a with adeno-associated virus (AAV) may improve calcium recycling and provide long-lasting benefits in DMD. To test this, we injected an AAV9 human SERCA2a vector (6 × 1012 viral genome particles/mouse) intravenously to 3-month-old mdx mice, the most commonly used DMD model. Immunostaining and western blot showed robust human SERCA2a expression in the heart and skeletal muscle for 18 months. Concomitantly, SR calcium uptake was significantly improved in these tissues. SERCA2a therapy significantly enhanced grip force and treadmill performance, completely prevented myocardial fibrosis, and normalized electrocardiograms (ECGs). Cardiac catheterization showed normalization of multiple systolic and diastolic hemodynamic parameters in treated mice. Importantly, chamber dilation was completely prevented, and ejection fraction was restored to the wild-type level. Our results suggest that a single systemic AAV9 SERCA2a therapy has the potential to provide long-lasting benefits for DMD.


Subject(s)
Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/therapy , Gene Expression , Genetic Therapy , Muscular Dystrophy, Duchenne/complications , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Administration, Intravenous , Animals , Dependovirus/genetics , Disease Models, Animal , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Sarcoplasmic Reticulum/metabolism , Time Factors , Transduction, Genetic
6.
Am J Physiol Cell Physiol ; 317(4): C813-C824, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31365291

ABSTRACT

Reduction in the expression of sarcolipin (SLN), an inhibitor of sarco(endo)plasmic reticulum (SR) Ca2+-ATPase (SERCA), ameliorates severe muscular dystrophy in mice. However, the mechanism by which SLN inhibition improves muscle structure remains unclear. Here, we describe the previously unknown function of SLN in muscle differentiation in Duchenne muscular dystrophy (DMD). Overexpression of SLN in C2C12 resulted in decreased SERCA pump activity, reduced SR Ca2+ load, and increased intracellular Ca2+ (Cai2+) concentration. In addition, SLN overexpression resulted in altered expression of myogenic markers and poor myogenic differentiation. In dystrophin-deficient dog myoblasts and myotubes, SLN expression was significantly high and associated with defective Cai2+ cycling. The dystrophic dog myotubes were less branched and associated with decreased autophagy and increased expression of mitochondrial fusion and fission proteins. Reduction in SLN expression restored these changes and enhanced dystrophic dog myoblast fusion during differentiation. In summary, our data suggest that SLN upregulation is an intrinsic secondary change in dystrophin-deficient myoblasts and could account for the Cai2+ mishandling, which subsequently contributes to poor myogenic differentiation. Accordingly, reducing SLN expression can improve the Cai2+ cycling and differentiation of dystrophic myoblasts. These findings provide cellular-level supports for targeting SLN expression as a therapeutic strategy for DMD.


Subject(s)
Calcium/metabolism , Muscle Development/physiology , Muscle Proteins/metabolism , Muscular Dystrophy, Duchenne/metabolism , Proteolipids/metabolism , Animals , Cell Differentiation/physiology , Dogs , Dystrophin/deficiency , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Myoblasts/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
7.
Am J Physiol Heart Circ Physiol ; 308(3): H240-9, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25485900

ABSTRACT

Inhibition of ß-adrenergic receptor (ß-AR) signaling is one of the most common therapeutic approaches for patients with arrhythmias. Adenylyl cyclase (AC) is the key enzyme responsible for transducing ß-AR stimulation to increases in cAMP. The two major AC isoforms in the heart are types 5 and 6. Therefore, it is surprising that prior studies on overexpression of AC5 and AC6 in transgenic (Tg) mice have not examined mediation of arrhythmogenesis. Our goal was to examine the proarrhythmic substrate in AC5Tg hearts. Intracellular calcium ion (Ca(2+) i) was imaged in fluo-4 AM-loaded ventricular myocytes. The sarcoplasmic reticulum (SR) Ca(2+) content, fractional Ca(2+) release, and twitch Ca(2+) transient were significantly higher in the AC5Tg vs. wild-type (WT) myocytes, indicating Ca(2+) overload in AC5Tg myocytes. Action potential (AP) duration was significantly longer in AC5Tg than in WT myocytes. Additionally, AC5Tg myocytes developed spontaneous Ca(2+) waves in a larger fraction compared with WT myocytes, particularly when cells were exposed to isoproterenol. The Ca(2+) waves further induced afterdepolarizations and triggered APs. AC5Tg hearts had increased level of SERCA2a, oxidized Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), and phosphorylation of ryanodine receptors (RyR) at the CaMKII site, especially after isoproterenol treatment. This was consistent with higher reactive oxygen species production in AC5Tg myocytes after isoproterenol treatment. Isoproterenol induced more severe arrhythmias in AC5Tg than in WT mice. We conclude that AC5 overexpression promotes arrhythmogenesis, by inducing SR Ca(2+) overload and hyperactivation of RyR (phosphorylation by CaMKII), which in turn induces spontaneous Ca(2+) waves and afterdepolarizations.


Subject(s)
Adenylyl Cyclases/metabolism , Arrhythmias, Cardiac/metabolism , Action Potentials , Adenylyl Cyclases/genetics , Animals , Calcium/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiotonic Agents/pharmacology , Cells, Cultured , Heart Ventricles/cytology , Heart Ventricles/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Isoproterenol/pharmacology , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
8.
Mol Ther Methods Clin Dev ; 32(2): 101268, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38911286

ABSTRACT

Excessive cytosolic calcium accumulation contributes to muscle degeneration in Duchenne muscular dystrophy (DMD). Sarco/endoplasmic reticulum calcium ATPase (SERCA) is a sarcoplasmic reticulum (SR) calcium pump that actively transports calcium from the cytosol into the SR. We previously showed that adeno-associated virus (AAV)-mediated SERCA2a therapy reduced cytosolic calcium overload and improved muscle and heart function in the murine DMD model. Here, we tested whether AAV SERCA2a therapy could ameliorate muscle disease in the canine DMD model. 7.83 × 1013 vector genome particles of the AAV vector were injected into the extensor carpi ulnaris (ECU) muscles of four juvenile affected dogs. Contralateral ECU muscles received excipient. Three months later, we observed widespread transgene expression and significantly increased SERCA2a levels in the AAV-injected muscles. Treatment improved SR calcium uptake, significantly reduced calpain activity, significantly improved contractile kinetics, and significantly enhanced resistance to eccentric contraction-induced force loss. Nonetheless, muscle histology was not improved. To evaluate the safety of AAV SERCA2a therapy, we delivered the vector to the ECU muscle of adult normal dogs. We achieved strong transgene expression without altering muscle histology and function. Our results suggest that AAV SERCA2a therapy has the potential to improve muscle performance in a dystrophic large mammal.

9.
JCI Insight ; 9(9)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38564291

ABSTRACT

Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease associated with cardiomyopathy. DMD cardiomyopathy is characterized by abnormal intracellular Ca2+ homeostasis and mitochondrial dysfunction. We used dystrophin and utrophin double-knockout (mdx:utrn-/-) mice in a sarcolipin (SLN) heterozygous-knockout (sln+/-) background to examine the effect of SLN reduction on mitochondrial function in the dystrophic myocardium. Germline reduction of SLN expression in mdx:utrn-/- mice improved cardiac sarco/endoplasmic reticulum (SR) Ca2+ cycling, reduced cardiac fibrosis, and improved cardiac function. At the cellular level, reducing SLN expression prevented mitochondrial Ca2+ overload, reduced mitochondrial membrane potential loss, and improved mitochondrial function. Transmission electron microscopy of myocardial tissues and proteomic analysis of mitochondria-associated membranes showed that reducing SLN expression improved mitochondrial structure and SR-mitochondria interactions in dystrophic cardiomyocytes. These findings indicate that SLN upregulation plays a substantial role in the pathogenesis of cardiomyopathy and that reducing SLN expression has clinical implications in the treatment of DMD cardiomyopathy.


Subject(s)
Cardiomyopathies , Dystrophin , Mice, Inbred mdx , Mice, Knockout , Muscle Proteins , Muscular Dystrophy, Duchenne , Proteolipids , Utrophin , Animals , Male , Mice , Calcium/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondria, Heart/genetics , Muscle Proteins/metabolism , Muscle Proteins/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proteolipids/metabolism , Proteolipids/genetics , Utrophin/genetics , Utrophin/metabolism
10.
Am J Physiol Cell Physiol ; 304(2): C194-206, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23135699

ABSTRACT

The role of SMA and SMB smooth muscle myosin heavy chain (MHC) isoforms in tonic and phasic contractions was studied in phasic (longitudinal ileum and stomach circular antrum) and tonic (stomach circular fundus) smooth muscle tissues of SMB knockout mice. Knocking out the SMB MHC gene eliminated SMB MHC protein expression and resulted in upregulation of the SMA MHC protein without altering the total MHC protein level. Switching from SMB to SMA MHC protein expression decreased the rate of the force transient and increased the sustained tonic force in SMB((-/-)) ileum and antrum with high potassium (KPSS) but not with carbachol (CCh) stimulation. The increased tonic contraction under the depolarized condition was not through changes in second messenger signaling pathways (PKC/CPI-17 or Rho/ROCK signaling pathway) or LC(20) phosphorylation. Biochemical analyses showed that the expression of contractile regulatory proteins (MLCK, MLCP, PKCδ, and CPI-17) did not change significantly in tissues tested except for PKCα protein expression being significantly decreased in the SMB((-/-)) antrum. However, specifically activating PKCα with phorbol dibutyrate (PDBu) was not significantly different in knockout and wild-type tissues, with total force being a fraction of the force generation with KPSS or CCh stimulation in SMB((-/-)) ileum and antrum. Taken together, these data show removing the SMB MHC protein expression with a compensatory increase in the SMA MHC protein results in enhanced sustained KPSS-induced tonic contraction with a reduced rate of force generation in these phasic tissues.


Subject(s)
Ileum/physiology , Muscle Contraction/physiology , Muscle Strength/physiology , Muscle, Smooth/physiology , Myosin Heavy Chains/physiology , Pyloric Antrum/physiology , Smooth Muscle Myosins/physiology , Animals , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Ileum/cytology , Ileum/drug effects , Mice , Mice, Knockout , Muscle Contraction/drug effects , Muscle Strength/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Potassium/pharmacology , Protein Kinase C-alpha/biosynthesis , Protein Kinase C-alpha/physiology , Pyloric Antrum/cytology , Pyloric Antrum/drug effects , Second Messenger Systems/drug effects , Second Messenger Systems/genetics , Second Messenger Systems/physiology , Smooth Muscle Myosins/biosynthesis , Smooth Muscle Myosins/genetics
11.
Am J Physiol Heart Circ Physiol ; 305(10): H1440-50, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24014679

ABSTRACT

Xanthine oxidase (XO) is increased in human and rat left ventricular (LV) myocytes with volume overload (VO) of mitral regurgitation and aortocaval fistula (ACF). In the setting of increased ATP demand, XO-mediated ROS can decrease mitochondrial respiration and contractile function. Thus, we tested the hypothesis that XO inhibition improves cardiomyocyte bioenergetics and LV function in chronic ACF in the rat. Sprague-Dawley rats were randomized to either sham or ACF ± allopurinol (100 mg·kg(-1)·day(-1), n ≥7 rats/group). Echocardiography at 8 wk demonstrated a similar 37% increase in LV end-diastolic dimension (P < 0.001), a twofold increase in LV end-diastolic pressure/wall stress (P < 0.05), and a twofold increase in lung weight (P < 0.05) in treated and untreated ACF groups versus the sham group. LV ejection fraction, velocity of circumferential shortening, maximal systolic elastance, and contractile efficiency were significantly depressed in ACF and significantly improved in ACF + allopurinol rats, all of which occurred in the absence of changes in the maximum O2 consumption rate measured in isolated cardiomyocytes using the extracellular flux analyzer. However, the improvement in contractile function is not paralleled by any attenuation in LV dilatation, LV end-diastolic pressure/wall stress, and lung weight. In conclusion, allopurinol improves LV contractile function and efficiency possibly by diminishing the known XO-mediated ROS effects on myofilament Ca(2+) sensitivity. However, LV remodeling and diastolic properties are not improved, which may explain the failure of XO inhibition to improve symptoms and hospitalizations in patients with severe heart failure.


Subject(s)
Allopurinol/pharmacology , Cardiotonic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Heart Failure/drug therapy , Heart Ventricles/drug effects , Myocytes, Cardiac/drug effects , Systole/drug effects , Ventricular Function, Left/drug effects , Xanthine Oxidase/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling/drug effects , Creatine Kinase/metabolism , Diastole/drug effects , Disease Models, Animal , Energy Metabolism/drug effects , Heart Failure/enzymology , Heart Failure/etiology , Heart Failure/physiopathology , Heart Ventricles/diagnostic imaging , Heart Ventricles/enzymology , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Myocytes, Cardiac/enzymology , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/enzymology , Stroke Volume/drug effects , Time Factors , Ultrasonography , Ventricular Pressure/drug effects , Xanthine Oxidase/metabolism
12.
J Muscle Res Cell Motil ; 34(5-6): 349-56, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23748997

ABSTRACT

Abnormal intracellular Ca(2+) handling is an important factor in the progressive functional decline of dystrophic muscle. In the present study, we investigated the function of sarco(endo)plasmic reticulum (SR) Ca(2+) ATPase (SERCA) in various dystrophic muscles of mouse models of Duchenne muscular dystrophy. Our studies show that the protein expression of sarcolipin, a key regulator of the SERCA pump is abnormally high and correlates with decreased maximum velocity of SR Ca(2+) uptake in the soleus, diaphragm and quadriceps of mild (mdx) and severe (mdx:utr-/-) dystrophic mice. These changes are more pronounced in the muscles of mdx:utr-/- mice. We also found increased expression of SERCA2a and calsequestrin specifically in the dystrophic quadriceps. Immunostaining analysis further showed that SERCA2a expression is associated both with fibers expressing slow-type myosin and regenerating fibers expressing embryonic myosin. Together, our data suggest that sarcolipin upregulation is a common secondary alteration in all dystrophic muscles and contributes to the abnormal elevation of intracellular Ca(2+) concentration via SERCA inhibition.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Proteolipids/biosynthesis , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
13.
bioRxiv ; 2023 Sep 13.
Article in English | MEDLINE | ID: mdl-37745514

ABSTRACT

Sympathetic nerves co-develop with their target organs and release neurotransmitters to stimulate their functions after maturation. Here, we provide the molecular mechanism that during sweat gland morphogenesis, neurotransmitters released from sympathetic nerves act first to promote sweat duct elongation via norepinephrine and followed by acetylcholine to specify sweat gland stem cell fate, which matches the sequence of neurotransmitter switch. Without neuronal signals during development, the basal cells switch to exhibit suprabasal (luminal) cell features. Sarcolipin (SLN), a key regulator of sarcoendoplasmic reticulum (SR) Ca 2+ -ATPase (SERCA), expression is significantly down-regulated in the sweat gland myoepithelial cells upon denervation. Loss of SLN in sweat gland myoepithelial cells leads to decreased intracellular Ca 2+ over time in response to ACh stimulation, as well as upregulation of luminal cell features. In cell culture experiments, we showed that contrary to the paradigm that elevation of Ca 2+ promote epidermal differentiation, specification of the glandular myoepithelial (basal) cells requires high Ca 2+ while lowering Ca 2+ level promotes luminal (suprabasal) cell fate. Our work highlights that neuronal signals not only act transiently for mature sweat glands to function, but also exert long-term impact on glandular stem cell specification through regulating intracellular Ca 2+ dynamics.

14.
J Am Heart Assoc ; 12(3): e027480, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36695318

ABSTRACT

Background Cardiomyopathy is a leading health threat in Duchenne muscular dystrophy (DMD). Cytosolic calcium upregulation is implicated in DMD cardiomyopathy. Calcium is primarily removed from the cytosol by the sarcoendoplasmic reticulum calcium ATPase (SERCA). SERCA activity is reduced in DMD. Improving SERCA function may treat DMD cardiomyopathy. Dwarf open reading frame (DWORF) is a recently discovered positive regulator for SERCA, hence, a potential therapeutic target. Methods and Results To study DWORF's involvement in DMD cardiomyopathy, we quantified DWORF expression in the heart of wild-type mice and the mdx model of DMD. To test DWORF gene therapy, we engineered and characterized an adeno-associated virus serotype 9-DWORF vector. To determine if this vector can mitigate DMD cardiomyopathy, we delivered it to 6-week-old mdx mice (6×1012 vector genome particles/mouse) via the tail vein. Exercise capacity, heart histology, and cardiac function were examined at 18 months of age. We found DWORF expression was significantly reduced at the transcript and protein levels in mdx mice. Adeno-associated virus serotype 9-DWORF vector significantly enhanced SERCA activity. Systemic adeno-associated virus serotype 9-DWORF therapy reduced myocardial fibrosis and improved treadmill running, electrocardiography, and heart hemodynamics. Conclusions Our data suggest that DWORF deficiency contributes to SERCA dysfunction in mdx mice and that DWORF gene therapy holds promise to treat DMD cardiomyopathy.


Subject(s)
Cardiomyopathies , Muscular Dystrophy, Duchenne , Mice , Animals , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Mice, Inbred mdx , Calcium , Open Reading Frames , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Genetic Therapy/methods
15.
Nat Commun ; 14(1): 602, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36746942

ABSTRACT

Polyglutamine expansion in the androgen receptor (AR) causes spinobulbar muscular atrophy (SBMA). Skeletal muscle is a primary site of toxicity; however, the current understanding of the early pathological processes that occur and how they unfold during disease progression remains limited. Using transgenic and knock-in mice and patient-derived muscle biopsies, we show that SBMA mice in the presymptomatic stage develop a respiratory defect matching defective expression of genes involved in excitation-contraction coupling (ECC), altered contraction dynamics, and increased fatigue. These processes are followed by stimulus-dependent accumulation of calcium into mitochondria and structural disorganization of the muscle triads. Deregulation of expression of ECC genes is concomitant with sexual maturity and androgen raise in the serum. Consistent with the androgen-dependent nature of these alterations, surgical castration and AR silencing alleviate the early and late pathological processes. These observations show that ECC deregulation and defective mitochondrial respiration are early but reversible events followed by altered muscle force, calcium dyshomeostasis, and dismantling of triad structure.


Subject(s)
Androgens , Bulbo-Spinal Atrophy, X-Linked , Mice , Animals , Androgens/metabolism , Bulbo-Spinal Atrophy, X-Linked/genetics , Calcium/metabolism , Muscle, Skeletal/metabolism , Receptors, Androgen/metabolism , Mitochondria/metabolism , Respiration , Disease Models, Animal
16.
Am J Physiol Cell Physiol ; 302(12): C1762-71, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22496245

ABSTRACT

Sarcolipin (SLN) is a key regulator of sarco(endo)plasmic reticulum (SR) Ca(2+)-ATPase (SERCA), and its expression is altered in diseased atrial myocardium. To determine the precise role of SLN in atrial Ca(2+) homeostasis, we developed a SLN knockout (sln-/-) mouse model and demonstrated that ablation of SLN enhances atrial SERCA pump activity. The present study is designed to determine the long-term effects of enhanced SERCA activity on atrial remodeling in the sln-/- mice. Calcium transient measurements show an increase in atrial SR Ca(2+) load and twitch Ca(2+) transients. Patch-clamping experiments demonstrate activation of the forward mode of sodium/calcium exchanger, increased L-type Ca(2+) channel activity, and prolongation of action potential duration at 90% repolarization in the atrial myocytes of sln-/- mice. Spontaneous Ca(2+) waves, delayed afterdepolarization, and triggered activities are frequent in the atrial myocytes of sln-/- mice. Furthermore, loss of SLN in atria is associated with increased interstitial fibrosis and altered expression of genes encoding collagen and other extracellular matrix proteins. Our results also show that the sln-/- mice are susceptible to atrial arrhythmias upon aging. Together, these findings indicate that ablation of SLN results in increased SERCA activity and SR Ca(2+) load, which, in turn, could cause abnormal intracellular Ca(2+) handling and atrial remodeling.


Subject(s)
Arrhythmias, Cardiac/metabolism , Atrial Function , Calcium Signaling , Muscle Proteins/deficiency , Myocytes, Cardiac/metabolism , Proteolipids/deficiency , Action Potentials , Aging/metabolism , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Calcium Channels, L-Type/metabolism , Collagen/metabolism , Fibrosis , Gene Expression Regulation , Genotype , Heart Atria/metabolism , Heart Atria/pathology , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Phenotype , Proteolipids/genetics , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Time Factors
17.
J Mol Cell Cardiol ; 50(1): 128-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21059353

ABSTRACT

Renin-angiotensin system inhibitors significantly reduce the incidence of arrhythmias. However, the underlying mechanism(s) is not well understood. We aim to test the hypothesis that angiotensin II (Ang II) induces early afterdepolarizations (EADs) and triggered activities (TAs) via the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-ROS-calmodulin kinase II (CaMKII) pathway. ROS production was analyzed in the isolated rabbit myocytes loaded with ROS dye. Ang II (1-2 µM) increased ROS fluorescence in myocytes, which was abolished by Ang II type 1 receptor blocker losartan, NADPH oxidase inhibitor apocynin, and antioxidant MnTMPyP, respectively. Action potentials were recorded using the perforated patch-clamp technique. EADs emerged in 27 out of 41 (66%) cells at 15.8 ± 1.6 min after Ang II (1-2 µM) perfusion. Ang II-induced EADs were eliminated by losartan, apocynin, or trolox. The CaMKII inhibitor KN-93 (n=6) and inhibitory peptide (AIP) (n=4) also suppressed Ang II-induced EADs, whereas the inactive analogue KN-92 did not. Nifedipine, a blocker of L-type Ca current (I(Ca)(2+)(,L)), or ranolazine, an inhibitor of late Na current (I(Na)(+)), abolished Ang II-induced EADs. The effects of Ang II on major membrane currents were evaluated using voltage clamp. While Ang II at same concentrations had no significant effect on total outward K(+) current, it enhanced I(Ca.L) and late I(Na), which were attenuated by losartan, apocynin, trolox, or KN-93. We conclude that Ang II induces EADs via intracellular ROS production through NADPH oxidase, activation of CaMKII, and enhancement of I(Ca,L) and late I(Na). These results provide evidence supporting a link between renin-angiotensin system and cardiac arrhythmias.


Subject(s)
Angiotensin II/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocytes, Cardiac/drug effects , Reactive Oxygen Species/metabolism , Acetanilides/pharmacology , Animals , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cells, Cultured , Electrophysiology , Heart Ventricles/cytology , Male , Nifedipine/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Rabbits , Ranolazine , Signal Transduction/drug effects , Sulfonamides/pharmacology
18.
Am J Physiol Cell Physiol ; 301(4): C841-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21697544

ABSTRACT

Sarcolipin (SLN) inhibits sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA) pumps. To evaluate the physiological significance of SLN in skeletal muscle, we compared muscle contractility and SERCA activity between Sln-null and wild-type mice. SLN protein expression in wild-type mice was abundant in soleus and red gastrocnemius (RG), low in extensor digitorum longus (EDL), and absent from white gastrocnemius (WG). SERCA activity rates were increased in soleus and RG, but not in EDL or WG, from Sln-null muscles, compared with wild type. No differences were seen between wild-type and Sln-null EDL muscles in force-frequency curves or maximum rates of force development (+dF/dt). Maximum relaxation rates (-dF/dt) of EDL were higher in Sln-null than wild type across a range of submaximal stimulation frequencies, but not during a twitch or peak tetanic contraction. For soleus, no differences were seen between wild type and Sln-null in peak tetanic force or +dF/dt; however, force-frequency curves showed that peak force during a twitch and 10-Hz contraction was lower in Sln-null. Changes in the soleus force-frequency curve corresponded with faster rates of force relaxation at nearly all stimulation frequencies in Sln-null compared with wild type. Repeated tetanic stimulation of soleus caused increased (-dF/dt) in wild type, but not in Sln-null. No compensatory responses were detected in analysis of other Ca(2+) regulatory proteins using Western blotting and immunohistochemistry or myosin heavy chain expression using immunofluorescence. These results show that 1) SLN regulates Ca(2+)-ATPase activity thereby regulating contractile kinetics in at least some skeletal muscles, 2) the functional significance of SLN is graded to the endogenous SLN expression level, and 3) SLN inhibitory effects on SERCA function are relieved in response to repeated contractions thus enhancing relaxation rates.


Subject(s)
Biological Transport/physiology , Calcium/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Relaxation/physiology , Proteolipids/genetics , Proteolipids/metabolism , Animals , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Muscle Contraction/physiology , Mutation , Myocardium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
19.
Biochem Biophys Res Commun ; 410(1): 97-101, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21640081

ABSTRACT

Sarcolipin (SLN), a key regulator of cardiac sarco(endo)plasmic reticulum (SR) Ca(2+) ATPase, is predominantly expressed in atria and mediates ß-adrenergic responses. Studies have shown that SLN mRNA expression is decreased in human chronic atrial fibrillation (AF) and in aortic banded mouse atria; however, SLN protein expression in human atrial pathology and its role in atrial SR Ca(2+) uptake are not yet elucidated. In the present study, we determined the expression of major SR Ca(2+) handling proteins in atria of human AF patients and in human and in a mouse model of heart failure (HF). We found that the expression of SR Ca(2+) uptake and Ca(2+) release channel proteins are significantly decreased in atria but not in the ventricles of pressure-overload induced HF in mice. In human AF and HF, the expression of SLN protein was significantly decreased; whereas the expressions of other major SR Ca(2+) handling proteins were not altered. Further, we found that the SR Ca(2+) uptake was significantly increased in human AF. The selective downregulation of SLN and enhanced SR Ca(2+) uptake in human AF suggest that SLN downregulation could play an important role in abnormal intracellular Ca(2+) cycling in atrial pathology.


Subject(s)
Atrial Fibrillation/metabolism , Calcium/metabolism , Muscle Proteins/biosynthesis , Proteolipids/biosynthesis , Sarcoplasmic Reticulum/metabolism , Adult , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Down-Regulation , Female , Heart Atria/metabolism , Humans , Ion Transport , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle Proteins/genetics , Proteolipids/genetics
20.
Proc Natl Acad Sci U S A ; 105(47): 18614-8, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-19011095

ABSTRACT

The physiological relevance of smooth muscle myosin isoforms SM1 and SM2 has not been understood. In this study we generated a mouse model specifically deficient in SM2 myosin isoform but expressing SM1, using an exon-specific gene targeting strategy. The SM2 homozygous knockout (SM2(-/-)) mice died within 30 days after birth, showing pathologies including segmental distention of alimentary tract, retention of urine in renal pelvis, distension of bladder, and the development of end-stage hydronephrosis. In contrast, the heterozygous (SM2(+/-)) mice appeared normal and reproduced well. In SM2(-/-) bladder smooth muscle the loss of SM2 myosin was accompanied by a concomitant down-regulation of SM1 and a reduced number of thick filaments. However, muscle strips from SM2(-/-) bladder showed increased contraction to K(+) depolarization or in response to M3 receptor agonist Carbachol. An increase of contraction was also observed in SM2(-/-) aorta. However, the SM2(-/-) bladder was associated with unaltered regulatory myosin light chain (MLC20) phosphorylation. Moreover, other contractile proteins, such as alpha-actin and tropomyosin, were not altered in SM2(-/-) bladder. Therefore, the loss of SM2 myosin alone could have induced hypercontractility in smooth muscle, suggesting that distinctly from SM1, SM2 may negatively modulate force development during smooth muscle contraction. Also, because SM2(-/-) mice develop lethal multiorgan dysfunctions, we propose this regulatory property of SM2 is essential for normal contractile activity in postnatal smooth muscle physiology.


Subject(s)
Smooth Muscle Myosins/physiology , Animals , Animals, Newborn , Carbachol/pharmacology , Female , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/drug effects , Phosphorylation , Smooth Muscle Myosins/genetics , Smooth Muscle Myosins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL