Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
EMBO J ; 38(24): e102578, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31381180

ABSTRACT

Exercise stimulates cellular and physiological adaptations that are associated with widespread health benefits. To uncover conserved protein phosphorylation events underlying this adaptive response, we performed mass spectrometry-based phosphoproteomic analyses of skeletal muscle from two widely used rodent models: treadmill running in mice and in situ muscle contraction in rats. We overlaid these phosphoproteomic signatures with cycling in humans to identify common cross-species phosphosite responses, as well as unique model-specific regulation. We identified > 22,000 phosphosites, revealing orthologous protein phosphorylation and overlapping signaling pathways regulated by exercise. This included two conserved phosphosites on stromal interaction molecule 1 (STIM1), which we validate as AMPK substrates. Furthermore, we demonstrate that AMPK-mediated phosphorylation of STIM1 negatively regulates store-operated calcium entry, and this is beneficial for exercise in Drosophila. This integrated cross-species resource of exercise-regulated signaling in human, mouse, and rat skeletal muscle has uncovered conserved networks and unraveled crosstalk between AMPK and intracellular calcium flux.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Proteomics/methods , Stromal Interaction Molecule 1/metabolism , Animals , Calcium Signaling/physiology , Drosophila , Female , Humans , Male , Membrane Proteins , Mice , Muscle, Skeletal/metabolism , Phosphorylation , Protein Conformation , Rats , Rats, Wistar , Signal Transduction , Stromal Interaction Molecule 1/chemistry , Stromal Interaction Molecule 1/genetics
2.
Exp Physiol ; 106(7): 1597-1611, 2021 07.
Article in English | MEDLINE | ID: mdl-33963617

ABSTRACT

NEW FINDINGS: What is the central question of this study? Striated muscle activator of rho signalling (STARS) is an actin-binding protein that regulates transcriptional pathways controlling muscle function, growth and myogenesis, processes that are impaired in dystrophic muscle: what is the regulation of the STARS pathway in Duchenne muscular dystrophy (DMD)? What is the main finding and its importance? Members of the STARS signalling pathway are reduced in the quadriceps of patients with DMD and in mouse models of muscular dystrophy. Overexpression of STARS in the dystrophic deficient mdx mouse model increased maximal isometric specific force and upregulated members of the actin cytoskeleton and oxidative phosphorylation pathways. Regulating STARS may be a therapeutic approach to enhance muscle health. ABSTRACT: Duchenne muscular dystrophy (DMD) is characterised by impaired cytoskeleton organisation, cytosolic calcium handling, oxidative stress and mitochondrial dysfunction. This results in progressive muscle damage, wasting and weakness and premature death. The striated muscle activator of rho signalling (STARS) is an actin-binding protein that activates the myocardin-related transcription factor-A (MRTFA)/serum response factor (SRF) transcriptional pathway, a pathway regulating cytoskeletal structure and muscle function, growth and repair. We investigated the regulation of the STARS pathway in the quadriceps muscle from patients with DMD and in the tibialis anterior (TA) muscle from the dystrophin-deficient mdx and dko (utrophin and dystrophin null) mice. Protein levels of STARS, SRF and RHOA were reduced in patients with DMD. STARS, SRF and MRTFA mRNA levels were also decreased in DMD muscle, while Stars mRNA levels were decreased in the mdx mice and Srf and Mrtfa mRNAs decreased in the dko mice. Overexpressing human STARS (hSTARS) in the TA muscles of mdx mice increased maximal isometric specific force by 13% (P < 0.05). This was not associated with changes in muscle mass, fibre cross-sectional area, fibre type, centralised nuclei or collagen deposition. Proteomics screening followed by pathway enrichment analysis identified that hSTARS overexpression resulted in 31 upregulated and 22 downregulated proteins belonging to the actin cytoskeleton and oxidative phosphorylation pathways. These pathways are impaired in dystrophic muscle and regulate processes that are vital for muscle function. Increasing the STARS protein in dystrophic muscle improves muscle force production, potentially via synergistic regulation of cytoskeletal structure and energy production.


Subject(s)
Muscular Dystrophy, Duchenne , Oxidative Phosphorylation , Actin Cytoskeleton/metabolism , Animals , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Humans , Mice , Mice, Inbred mdx , Microfilament Proteins , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism
3.
Int J Obes (Lond) ; 44(2): 500-509, 2020 02.
Article in English | MEDLINE | ID: mdl-30705392

ABSTRACT

BACKGROUND: Obesity is associated with development of insulin resistance in adipose tissue (AT). Human obesity has been associated with increased glycogen deposition in adipocytes. Adipocytes synthesise glycogen prior to the formation of lipids. The present study examined adipose glycogen content in obese Zucker rats and the effect of fasting on glycogen-metabolising enzymes. We hypothesised that obesity imposes a blunted response to fasting through impaired activation of glycogen-metabolizing enzymes, which dampens glycogen mobilization in obese Zucker rats. METHODS: We investigated the effect of 24h fasting on AT glycogen metabolism in 12-week old obese Zucker rats. Epididymal fat pads were collected from rats fed ad-libitum and fasted for 24h. Glycogen content, glycogen synthase and phosphorylase enzyme activity, and PKA activity were analysed as well as total and phosphorylated protein content for glycogen-metabolizing enzymes glycogen synthase and phosphorylase, glucose transporter GLUT4, and cAMP-dependent response element binding protein levels. RESULTS: Twelve-week old obese Zucker rats showed increased AT glycogen content (adipose glycogen content [mean ± SD], lean: 3.95 ± 2.78 to 0.75 + 0.69 µg.mg-1; p < 0.005 fed vs fasted, and obese: 5.23 ± 3.38 to 5.019 ± 1.99 µg.mg-1; p = ns fed and fasted and p < 0.005 lean vs obese), and impaired fasting-induced glycogen mobilization following a 24h fast. These defects were associated with dysfunctional glycogen-metabolizing enzymes, characterized by: (1) blunted phosphorylation-mediated activation and downregulated protein expression of glycogen phosphorylase, and (2) an impaired phosphorylation-mediated inactivation of glycogen synthase. Furthermore, these defects were related to impaired fasting-induced protein kinase A (PKA) activation. CONCLUSION: This study provides evidence of a defective glycogen metabolism in the adipose associated with impaired fasting-induced activation of the upstream kinase protein kinase A, which render a converging point to obesity-related primary alterations in carbohydrate and lipid metabolism in the AT.


Subject(s)
Adipose Tissue/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Fasting/physiology , Glycogen/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Female , Insulin/metabolism , Male , Rats , Rats, Zucker
5.
Hum Mol Genet ; 23(25): 6697-711, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25082828

ABSTRACT

Mutations in dystrophin lead to Duchenne muscular dystrophy, which is among the most common human genetic disorders. Dystrophin nucleates assembly of the dystrophin-glycoprotein complex (DGC), and a defective DGC disrupts an essential link between the intracellular cytoskeleton and the basal lamina, leading to progressive muscle wasting. In vitro studies have suggested that dystrophin phosphorylation may affect interactions with actin or syntrophin, yet whether this occurs in vivo or affects protein function remains unknown. Utilizing nanoflow liquid chromatography mass spectrometry, we identified 18 phosphorylated residues within endogenous dystrophin. Mutagenesis revealed that phosphorylation at S3059 enhances the dystrophin-dystroglycan interaction and 3D modeling utilizing the Rosetta software program provided a structural model for how phosphorylation enhances this interaction. These findings demonstrate that phosphorylation is a key mechanism regulating the interaction between dystrophin and the DGC and reveal that posttranslational modification of a single amino acid directly modulates the function of dystrophin.


Subject(s)
Dystroglycans/metabolism , Dystrophin-Associated Proteins/metabolism , Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Cell Line , Cysteine/chemistry , Cysteine/metabolism , Dystroglycans/chemistry , Dystroglycans/genetics , Dystrophin/chemistry , Dystrophin/genetics , Dystrophin-Associated Proteins/chemistry , Dystrophin-Associated Proteins/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Myoblasts/cytology , Myoblasts/metabolism , Phosphorylation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/chemistry , Serine/metabolism , Signal Transduction
6.
J Neurosci Res ; 94(5): 389-400, 2016 May.
Article in English | MEDLINE | ID: mdl-26864466

ABSTRACT

This study seeks to determine whether knockdown of basal forebrain p75 neurotrophin receptor (p75(NTR) ) expression elicits increased hippocampal choline acetyltransferase (ChAT) activity in mature animals. Antisense (AS) oligonucleotides (oligos) targeting p75(NTR) were infused into the medial septal area of mature rats continuously for 4 weeks. In all rats, the cannula outlet was placed equidistant between the left and the right sides of the vertical diagonal band of Broca. We tested phosphorothioate (PS), morpholino (Mo), and gapmer (mixed PS/RNA) oligos. Gapmer AS infusions of 7.5 and 22 µg/day decreased septal p75(NTR) mRNA by 34% and 48%, respectively. The same infusions increased hippocampal ChAT activity by 41% and 55%. Increased hippocampal ChAT activity correlated strongly with septal p75(NTR) downregulation in individual rats. Infusions of PS and Mo AS oligos did not downregulate p75(NTR) mRNA or stimulate ChAT activity. These results demonstrate that p75(NTR) can dynamically regulate hippocampal ChAT activity in the mature CNS. They also reveal the different efficacies of three diverse AS oligo chemistries when infused intracerebrally. Among the three types, gapmer oligos worked best.


Subject(s)
Basal Forebrain/metabolism , Choline O-Acetyltransferase/metabolism , Gene Knockdown Techniques/methods , Hippocampus/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Choline O-Acetyltransferase/genetics , Enzyme Activation/physiology , Female , Nerve Tissue Proteins , Rats , Receptors, Growth Factor , Receptors, Nerve Growth Factor/genetics
7.
Exp Physiol ; 99(4): 675-87, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24443351

ABSTRACT

New Findings What is the central question of this study? The Notch signalling pathway plays an important role in muscle regeneration, and activation of the pathway has been shown to enhance muscle regeneration in aged mice. It is unknown whether Notch activation will have a similarly beneficial effect on muscle regeneration in the context of Duchenne muscular dystrophy (DMD). What is the main finding and its importance? Although expression of Notch signalling components is altered in both mouse models of DMD and in human DMD patients, activation of the Notch signalling pathway does not confer any functional benefit on muscles from dystrophic mice, suggesting that other signalling pathways may be more fruitful targets for manipulation in treating DMD. Abstract In Duchenne muscular dystrophy (DMD), muscle damage and impaired regeneration lead to progressive muscle wasting, weakness and premature death. The Notch signalling pathway represents a central regulator of gene expression and is critical for cellular proliferation, differentiation and apoptotic signalling during all stages of embryonic muscle development. Notch activation improves muscle regeneration in aged mice, but its potential to restore regeneration and function in muscular dystrophy is unknown. We performed a comprehensive examination of several genes involved in Notch signalling in muscles from dystrophin-deficient mdx and dko (utrophin- and dystrophin-null) mice and DMD patients. A reduction of Notch1 and Hes1 mRNA in tibialis anterior muscles of dko mice and quadriceps muscles of DMD patients and a reduction of Hes1 mRNA in the diaphragm of the mdx mice were observed, with other targets being inconsistent across species. Activation and inhibition of Notch signalling, followed by measures of muscle regeneration and function, were performed in the mouse models of DMD. Notch activation had no effect on functional regeneration in C57BL/10, mdx or dko mice. Notch inhibition significantly depressed the frequency-force relationship in regenerating muscles of C57BL/10 and mdx mice after injury, indicating reduced force at each stimulation frequency, but enhanced the frequency-force relationship in muscles from dko mice. We conclude that while Notch inhibition produces slight functional defects in dystrophic muscle, Notch activation does not significantly improve muscle regeneration in murine models of muscular dystrophy. Furthermore, the inconsistent expression of Notch targets between murine models and DMD patients suggests caution when making interspecies comparisons.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Muscular Dystrophy, Duchenne/metabolism , Receptors, Notch/metabolism , Signal Transduction , Adolescent , Adult , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biopsy , Case-Control Studies , Child , Child, Preschool , Disease Models, Animal , Dystrophin/deficiency , Dystrophin/genetics , Elapid Venoms , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Infant , Mice, Inbred mdx , Mice, Knockout , Muscle Contraction , Muscle Development , Muscle Strength , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Diseases/chemically induced , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , RNA, Messenger/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Notch/genetics , Regeneration , Transcription Factor HES-1 , Utrophin/deficiency , Utrophin/genetics , Young Adult
8.
Dis Model Mech ; 17(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38602028

ABSTRACT

Duchenne muscular dystrophy (DMD) is a devastating monogenic skeletal muscle-wasting disorder. Although many pharmacological and genetic interventions have been reported in preclinical studies, few have progressed to clinical trials with meaningful benefit. Identifying therapeutic potential can be limited by availability of suitable preclinical mouse models. More rigorous testing across models with varied background strains and mutations can identify treatments for clinical success. Here, we report the generation of a DMD mouse model with a CRISPR-induced deletion within exon 62 of the dystrophin gene (Dmd) and the first generated in BALB/c mice. Analysis of mice at 3, 6 and 12 months of age confirmed loss of expression of the dystrophin protein isoform Dp427 and resultant dystrophic pathology in limb muscles and the diaphragm, with evidence of centrally nucleated fibers, increased inflammatory markers and fibrosis, progressive decline in muscle function, and compromised trabecular bone development. The BALB/c.mdx62 mouse is a novel model of DMD with associated variations in the immune response and muscle phenotype, compared with those of existing models. It represents an important addition to the preclinical model toolbox for developing therapeutic strategies.


Subject(s)
Disease Models, Animal , Dystrophin , Mice, Inbred BALB C , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Animals , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/genetics , Dystrophin/metabolism , Dystrophin/genetics , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Mice, Inbred mdx , Mice , Exons/genetics , Male , Fibrosis , Phenotype
9.
Int J Cancer ; 133(5): 1234-46, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23436228

ABSTRACT

Cancer cachexia describes the progressive skeletal muscle wasting and weakness associated with many cancers. Cachexia reduces mobility and quality of life and accounts for 20-30% of all cancer-related deaths. Activation of the renin-angiotensin system causes skeletal muscle wasting and weakness. We tested the hypothesis that treatment with the angiotensin converting enzyme (ACE) inhibitor, perindopril, would enhance whole body and skeletal muscle function in cachectic mice bearing Colon-26 (C-26) tumors. CD2F1 mice received a subcutaneous injection of phosphate buffered saline or C-26 tumor cells inducing either a mild or severe cachexia. The following day, one cohort of C-26 mice began receiving perindopril in their drinking water (4 mg kg(-1) day(-1) ) for 21 days. In mild and severe cachexia, perindopril increased measures of whole body function (grip strength and rotarod) and reduced fatigue in isolated contracting diaphragm muscle strips (p < 0.05). In severely cachectic mice, perindopril reduced tumor growth, improved locomotor activity and reduced fatigue of tibialis anterior muscles in situ (p < 0.05), which was associated with increased oxidative enzyme capacity (succinate deyhydrogenase, p < 0.05). Perindopril attenuated the increase in MuRF-1 and IL-6 mRNA expression and enhanced Akt phosphorylation in severely cachectic mice but neither body nor muscle mass was increased. These findings support the therapeutic potential of ACE inhibition for enhancing whole body function and reducing fatigue of respiratory muscles in early and late stage cancer cachexia and should be confirmed in future clinical trials. Since ACE inhibition alone did not enhance body or muscle mass, co-treatment with an anabolic agent may be required to address these aspects of cancer cachexia.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cachexia/drug therapy , Neoplasms/complications , Perindopril/pharmacology , Animals , Cachexia/metabolism , Cell Line, Tumor , Interleukin-6/genetics , Male , Mice , Motor Activity/drug effects , Muscle Fatigue/drug effects , Muscle Proteins/genetics , Muscle, Skeletal/drug effects , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
10.
Am J Physiol Regul Integr Comp Physiol ; 303(3): R321-31, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22673782

ABSTRACT

The role of the renin-angiotensin system (RAS) in vasoregulation is well established, but a localized RAS exists in multiple tissues and exerts diverse functions including autonomic control and thermogenesis. The role of the RAS in the maintenance and function of skeletal muscle is not well understood, especially the role of angiotensin peptides, which appear to contribute to muscle atrophy. We tested the hypothesis that mice lacking the angiotensin type 1A receptor (AT(1A)(-/-)) would exhibit enhanced whole body and skeletal muscle function and improved regeneration after severe injury. Despite 18- to 20-wk-old AT(1A)(-/-) mice exhibiting reduced muscle mass compared with controls (P < 0.05), the tibialis anterior (TA) muscles produced a 25% higher maximum specific (normalized) force (P < 0.05). Average fiber cross-sectional area (CSA) and fiber oxidative capacity was not different between groups, but TA muscles from AT(1A)(-/-) mice had a reduced number of muscle fibers as well as a higher proportion of type IIx/b fibers and a lower proportion of type IIa fibers (P < 0.05). Measures of whole body function (grip strength, rotarod performance, locomotor activity) were all improved in AT(1A)(-/-) mice (P < 0.05). Surprisingly, the recovery of muscle mass and fiber CSA following myotoxic injury was impaired in AT(1A)(-/-) mice, in part by impaired myoblast fusion, prolonged collagen infiltration and inflammation, and delayed expression of myogenic regulatory factors. The findings support the therapeutic potential of RAS inhibition for enhancing whole body and skeletal muscle function, but they also reveal the importance of RAS signaling in the maintenance of muscle mass and for normal fiber repair after injury.


Subject(s)
Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology , Receptor, Angiotensin, Type 1/deficiency , Renin-Angiotensin System/physiology , Wound Healing/physiology , Animals , Collagen Type II/physiology , Elapid Venoms/adverse effects , Elapid Venoms/pharmacology , Male , Mice , Mice, Knockout , Models, Animal , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/physiology , Muscle Strength/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscular Atrophy/pathology , Receptor, Angiotensin, Type 1/genetics , Signal Transduction/physiology
11.
Am J Pathol ; 176(1): 29-33, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19959813

ABSTRACT

The lack of functional dystrophin protein in Duchenne muscular dystrophy (DMD) renders muscle fibers highly fragile and susceptible to damage during contractions. Contraction-mediated injury is a major contributor to the progressive degeneration and etiology of muscle wasting in DMD. The prevailing understanding is that large fibers are highly susceptible to contraction damage and are affected preferentially, whereas smaller fibers are relatively spared in DMD. We tested the hypothesis that a pharmacological treatment that caused myofiber hypertrophy would increase the susceptibility of muscles from dystrophin-deficient mdx mice to contraction-induced injury, and thus aggravate the dystrophic pathology. The beta-agonist formoterol (100 microg/kg per day, i.p.) was administered to mdx mice for 28 days. Formoterol increased muscle mass, fiber cross-sectional area, and maximum force producing capacity by 30%, 23%, and 21%, respectively, in fast-twitch tibialis anterior muscles of mdx mice. Myofiber hypertrophy and increased maximum force producing capacity were also observed in the predominantly slow-twitch soleus muscles of mdx mice. Our original hypothesis was rejected since tibialis anterior muscles from formoterol-treated mdx mice had lower cumulative force deficits, indicating that they were less susceptible to contraction-induced injury. Formoterol treatment did not affect injury susceptibility in soleus muscles. These findings indicate that making dystrophic muscles bigger protects them from contraction damage and does not aggravate the dystrophic pathophysiology. These novel results further support the contention that anabolic agents have therapeutic potential for muscle wasting conditions including DMD.


Subject(s)
Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Animals , Ethanolamines/pharmacology , Formoterol Fumarate , Mice , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle Fibers, Fast-Twitch/drug effects , Organ Size/drug effects
12.
Am J Physiol Regul Integr Comp Physiol ; 301(3): R716-26, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21677277

ABSTRACT

Cancer cachexia describes the progressive skeletal muscle wasting and weakness in many cancer patients and accounts for >20% of cancer-related deaths. We tested the hypothesis that antibody-directed myostatin inhibition would attenuate the atrophy and loss of function in muscles of tumor-bearing mice. Twelve-week-old C57BL/6 mice received a subcutaneous injection of saline (control) or Lewis lung carcinoma (LLC) tumor cells. One week later, mice received either once weekly injections of saline (control, n = 12; LLC, n = 9) or a mouse chimera of anti-human myostatin antibody (PF-354, 10 mg·kg⁻¹·wk⁻¹, LLC+PF-354, n = 11) for 5 wk. Injection of LLC cells reduced muscle mass and maximum force of tibialis anterior (TA) muscles by 8-10% (P < 0.05), but the muscle atrophy and weakness were prevented with PF-354 treatment (P > 0.05). Maximum specific (normalized) force of diaphragm muscle strips was reduced with LLC injection (P < 0.05) but was not improved with PF-354 treatment (P > 0.05). PF-354 enhanced activity of oxidative enzymes in TA and diaphragm muscles of tumor-bearing mice by 118% and 89%, respectively (P < 0.05). Compared with controls, apoptosis that was not of myofibrillar or satellite cell origin was 140% higher in TA muscle cross sections from saline-treated LLC tumor-bearing mice (P < 0.05) but was not different in PF-354-treated tumor-bearing mice (P > 0.05). Antibody-directed myostatin inhibition attenuated the skeletal muscle atrophy and loss of muscle force-producing capacity in a murine model of cancer cachexia, in part by reducing apoptosis. The improvements in limb muscle mass and function highlight the therapeutic potential of antibody-directed myostatin inhibition for cancer cachexia.


Subject(s)
Antibodies/pharmacology , Cachexia/drug therapy , Carcinoma, Lewis Lung/complications , Muscle Weakness/drug therapy , Muscle, Skeletal/drug effects , Muscular Atrophy/drug therapy , Myostatin/antagonists & inhibitors , Analysis of Variance , Animals , Apoptosis/drug effects , Cachexia/etiology , Cachexia/metabolism , Cachexia/pathology , Cachexia/physiopathology , Carcinoma, Lewis Lung/pathology , Cell Proliferation/drug effects , Energy Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle Strength/drug effects , Muscle Weakness/etiology , Muscle Weakness/metabolism , Muscle Weakness/pathology , Muscle Weakness/physiopathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Myostatin/immunology , Myostatin/metabolism , Oxidation-Reduction , Phosphorylation , Smad3 Protein/metabolism , Succinate Dehydrogenase/metabolism , Time Factors
13.
Am J Physiol Regul Integr Comp Physiol ; 301(5): R1358-66, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21849632

ABSTRACT

Nitric oxide (NO) is an important signaling molecule produced in skeletal muscle primarily via the neuronal subtype of NO synthase (NOS1, or nNOS). While many studies have reported NO production to be important in muscle regeneration, none have examined the contribution of nNOS-derived NO to functional muscle regeneration (i.e., restoration of the muscle's ability to produce force) after acute myotoxic injury. In the present study, we tested the hypothesis that genetic deletion of nNOS would impair functional muscle regeneration after myotoxic injury in nNOS(-/-) mice. We found that nNOS(-/-) mice had lower body mass, lower muscle mass, and smaller myofiber cross-sectional area and that their tibialis anterior (TA) muscles produced lower absolute tetanic forces than those of wild-type littermate controls but that normalized or specific force was identical between the strains. In addition, muscles from nNOS(-/-) mice were more resistant to fatigue than those of wild-type littermates (P < 0.05). To determine whether deletion of nNOS affected muscle regeneration, TA muscles from nNOS(-/-) mice and wild-type littermates were injected with the myotoxin notexin to cause complete fiber degeneration, and muscle structure and function were assessed at 7 and 10 days postinjury. Myofiber cross-sectional area was lower in regenerating nNOS(-/-) mice than wild-type controls at 7 and 10 days postinjury; however, contrary to our original hypothesis, no difference in force-producing capacity of the TA muscle was evident between the two groups at either time point. Our findings reveal that nNOS is not essential for functional muscle regeneration after acute myotoxic damage.


Subject(s)
Isometric Contraction , Muscle Development , Muscle Strength , Muscle, Skeletal/enzymology , Muscular Diseases/enzymology , Nitric Oxide Synthase Type I/deficiency , Regeneration , Animals , Disease Models, Animal , Elapid Venoms , Electric Stimulation , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fatigue , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Diseases/chemically induced , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Nitric Oxide Synthase Type I/genetics , RNA, Messenger/metabolism , Recovery of Function , Time Factors
14.
FASEB J ; 24(11): 4433-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20624929

ABSTRACT

Sarcopenia is the progressive loss of skeletal muscle mass and function with advancing age, leading to reduced mobility and quality of life. We tested the hypothesis that antibody-directed myostatin inhibition would attenuate the decline in mass and function of muscles of aged mice and that apoptosis would be reduced. Eighteen-month-old C57BL/6 mice were treated for 14 wk with a once-weekly injection of saline (control, n=9) or a mouse chimera of anti-human myostatin antibody (PF-354, 10 mg/kg; n=12). PF-354 prevented the age-related reduction in body mass and increased soleus, gastrocnemius, and quadriceps muscle mass (P<0.05). PF-354 increased fiber cross-sectional area by 12% and enhanced maximum in situ force of tibialis anterior (TA) muscles by 35% (P<0.05). PF-354 increased the proportion of type IIa fibers by 114% (P<0.01) and enhanced activity of oxidative enzymes (SDH) by 39% (P<0.01). PF-354 reduced markers of apoptosis in TA muscle cross-sections by 56% (P<0.03) and reduced caspase3 mRNA by 65% (P<0.04). Antibody-directed myostatin inhibition attenuated the decline in mass and function of muscles of aging mice, in part, by reducing apoptosis. These observations identify novel roles for myostatin in regulation of muscle mass and highlight the therapeutic potential of antibody-directed myostatin inhibition for sarcopenia.


Subject(s)
Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Myostatin/antagonists & inhibitors , Signal Transduction , Aging/pathology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , Apoptosis , Humans , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Oxidoreductases/metabolism
15.
Nutrients ; 13(12)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34960110

ABSTRACT

Gastrointestinal (GI) dysfunction is an important, yet understudied condition associated with Duchenne muscular dystrophy (DMD), with patients reporting bloating, diarrhea, and general discomfort, contributing to a reduced quality of life. In the mdx mouse, the most commonly used mouse model of DMD, studies have confirmed GI dysfunction (reported as altered contractility and GI transit through the small and large intestine), associated with increased local and systemic inflammation. Sulforaphane (SFN) is a natural isothiocyanate with anti-inflammatory and anti-oxidative properties via its activation of Nrf2 signalling that has been shown to improve aspects of the skeletal muscle pathology in dystrophic mice. Whether SFN can similarly improve GI function in muscular dystrophy was unknown. Video imaging and spatiotemporal mapping to assess gastrointestinal contractions in isolated colon preparations from mdx and C57BL/10 mice revealed that SFN reduced contraction frequency when administered ex vivo, demonstrating its therapeutic potential to improve GI function in DMD. To confirm this in vivo, four-week-old male C57BL/10 and mdx mice received vehicle (2% DMSO/corn oil) or SFN (2 mg/kg in 2% DMSO/corn oil) via daily oral gavage five days/week for 4 weeks. SFN administration reduced fibrosis in the diaphragm of mdx mice but did not affect other pathological markers. Gene and protein analysis revealed no change in Nrf2 protein expression or activation of Nrf2 signalling after SFN administration and oral SFN supplementation did not improve GI function in mdx mice. Although ex vivo studies demonstrate SFN's therapeutic potential for reducing colon contractions, in vivo studies should investigate higher doses and/or alternate routes of administration to confirm SFN's potential to improve GI function in DMD.


Subject(s)
Gastrointestinal Diseases/drug therapy , Isothiocyanates/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Sulfoxides/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Colon/pathology , Diaphragm/pathology , Disease Models, Animal , Fibrosis/metabolism , Gastrointestinal Diseases/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , NF-E2-Related Factor 2/metabolism
16.
Mol Metab ; 45: 101157, 2021 03.
Article in English | MEDLINE | ID: mdl-33359740

ABSTRACT

OBJECTIVES: Preferential damage to fast, glycolytic myofibers is common in many muscle-wasting diseases, including Duchenne muscular dystrophy (DMD). Promoting an oxidative phenotype could protect muscles from damage and ameliorate the dystrophic pathology with therapeutic relevance, but developing efficacious strategies requires understanding currently unknown biological roles for dystrophin and utrophin in dystrophic muscle adaptation and plasticity. METHODS: Combining whole transcriptome RNA sequencing and mitochondrial proteomics with assessments of metabolic and contractile function, we investigated the roles of dystrophin and utrophin in fast-to-slow muscle remodeling with low-frequency electrical stimulation (LFS, 10 Hz, 12 h/d, 7 d/wk, 28 d) in mdx (dystrophin null) and dko (dystrophin/utrophin null) mice, two established preclinical models of DMD. RESULTS: Novel biological roles in adaptation were demonstrated by impaired transcriptional activation of estrogen-related receptor alpha-responsive genes supporting oxidative phosphorylation in dystrophic muscles. Further, utrophin expression in dystrophic muscles was required for LFS-induced remodeling of mitochondrial respiratory chain complexes, enhanced fiber respiration, and conferred protection from eccentric contraction-mediated damage. CONCLUSIONS: These findings reveal novel roles for dystrophin and utrophin during LFS-induced metabolic remodeling of dystrophic muscle and highlight the therapeutic potential of LFS to ameliorate the dystrophic pathology and protect from contraction-induced injury with important implications for DMD and related muscle disorders.


Subject(s)
Adaptation, Physiological/physiology , Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Utrophin/metabolism , Animals , Dystrophin/genetics , Male , Metabolic Engineering , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mitochondria/metabolism , Muscle Contraction , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Utrophin/genetics
17.
Genome Biol ; 22(1): 310, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34763716

ABSTRACT

A modified Chromium 10x droplet-based protocol that subsamples cells for both short-read and long-read (nanopore) sequencing together with a new computational pipeline (FLAMES) is developed to enable isoform discovery, splicing analysis, and mutation detection in single cells. We identify thousands of unannotated isoforms and find conserved functional modules that are enriched for alternative transcript usage in different cell types and species, including ribosome biogenesis and mRNA splicing. Analysis at the transcript level allows data integration with scATAC-seq on individual promoters, improved correlation with protein expression data, and linked mutations known to confer drug resistance to transcriptome heterogeneity.


Subject(s)
Nanopore Sequencing/methods , Protein Isoforms/genetics , Protein Isoforms/metabolism , Alternative Splicing , Animals , Exons , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Humans , Mice , RNA Splicing , RNA, Messenger , Transcriptome
18.
J Neurogastroenterol Motil ; 26(1): 133-146, 2020 Jan 30.
Article in English | MEDLINE | ID: mdl-31715094

ABSTRACT

BACKGROUND/AIMS: Patients with Duchenne muscular dystrophy exhibit significant, ongoing impairments in gastrointestinal (GI) function likely resulting from dysregulated nitric oxide production. Compounds increasing neuronal nitric oxide synthase expression and/or activity could improve GI dysfunction and enhance quality of life for dystrophic patients. We used video imaging and spatiotemporal mapping to identify GI dysfunction in mdx dystrophic mice and determine whether dietary intervention to enhance nitric oxide could alleviate aberrant colonic activity in muscular dystrophy. METHODS: Four-week-old male C57BL/10 and mdx mice received a specialized diet either with no supplementation (control) or supplemented (1 g/kg/day) with L-alanine, L-arginine, or L-citrulline for 8 weeks. At the conclusion of treatment, mice were sacrificed by cervical dislocation and colon motility examined by spatiotemporal (ST) mapping ex vivo. RESULTS: ST mapping identified increased contraction number in the mid and distal colon of mdx mice on control and L-alanine supplemented diets relative to C57BL/10 mice (P < 0.05). Administration of either L-arginine or L-citrulline attenuated contraction number in distal colons of mdx mice relative to C57BL/10 mice. CONCLUSIONS: GI dysfunction in Duchenne muscular dystrophy has been sadly neglected as an issue affecting quality of life. ST mapping identified regional GI dysfunction in the mdx dystrophic mouse. Dietary interventions to increase nitric oxide signaling in the GI tract reduced the number of colonic contractions and alleviated colonic constriction at rest. These findings in mdx mice reveal that L-arginine can improve colonic motility and has potential therapeutic relevance for alleviating GI discomfort, improving clinical care, and enhancing quality of life in Duchenne muscular dystrophy.

19.
PLoS One ; 14(2): e0212880, 2019.
Article in English | MEDLINE | ID: mdl-30811469

ABSTRACT

Muscles of older animals are more susceptible to injury and regenerate poorly, in part due to a persistent inflammatory response. The janus kinase (Jak)/signal transducer and activator of transcription (Stat) pathway mediates inflammatory signaling and is tightly regulated by the suppressor of cytokine signaling (SOCS) proteins, especially SOCS3. SOCS3 expression is altered in the muscle of aged animals and may contribute to the persistent inflammation and impaired regeneration. To test this hypothesis, we performed myotoxic injuries on mice with a tamoxifen-inducible deletion of SOCS3 specifically within the muscle stem cell compartment. Muscle stem cell-specific SOCS3 deletion reduced muscle mass at 14 days post-injury (-14%, P < 0.01), altered the myogenic transcriptional program, and reduced myogenic fusion based on the number of centrally-located nuclei per muscle fiber. Despite the delay in myogenesis, muscles with a muscle stem cell-specific deletion of SOCS3 were still able to regenerate after a single bout or multiple bouts of myotoxic injury. A reduction in SOCS3 expression in muscle stem cells is unlikely to be responsible for the incomplete muscle repair in aged animals.


Subject(s)
Aging/genetics , Gene Deletion , Regeneration/drug effects , Stem Cells/cytology , Suppressor of Cytokine Signaling 3 Protein/genetics , Tamoxifen/adverse effects , Aging/metabolism , Animals , Cells, Cultured , Female , Male , Mice , Muscle Cells/drug effects , Muscle Cells/physiology , PAX7 Transcription Factor/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism
20.
Sci Rep ; 9(1): 12982, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506484

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked genetic disease characterized by progressive muscle wasting and weakness and premature death. Glucocorticoids (e.g. prednisolone) remain the only drugs with a favorable impact on DMD patients, but not without side effects. We have demonstrated that glycine preserves muscle in various wasting models. Since glycine effectively suppresses the activity of pro-inflammatory macrophages, we investigated the potential of glycine treatment to ameliorate the dystrophic pathology. Dystrophic mdx and dystrophin-utrophin null (dko) mice were treated with glycine or L-alanine (amino acid control) for up to 15 weeks and voluntary running distance (a quality of life marker and strong correlate of lifespan in dko mice) and muscle morphology were assessed. Glycine increased voluntary running distance in mdx mice by 90% (P < 0.05) after 2 weeks and by 60% (P < 0.01) in dko mice co-treated with prednisolone over an 8 week treatment period. Glycine treatment attenuated fibrotic deposition in the diaphragm by 28% (P < 0.05) after 10 weeks in mdx mice and by 22% (P < 0.02) after 14 weeks in dko mice. Glycine treatment augmented the prednisolone-induced reduction in fibrosis in diaphragm muscles of dko mice (23%, P < 0.05) after 8 weeks. Our findings provide strong evidence that glycine supplementation may be a safe, simple and effective adjuvant for improving the efficacy of prednisolone treatment and improving the quality of life for DMD patients.


Subject(s)
Disease Models, Animal , Glycine Agents/administration & dosage , Glycine/administration & dosage , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Prednisolone/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology
SELECTION OF CITATIONS
SEARCH DETAIL