Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 1439, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35301320

ABSTRACT

During aging, the regenerative capacity of muscle stem cells (MuSCs) decreases, diminishing the ability of muscle to repair following injury. We found that the ability of MuSCs to regenerate is regulated by the primary cilium, a cellular protrusion that serves as a sensitive sensory organelle. Abolishing MuSC cilia inhibited MuSC proliferation in vitro and severely impaired injury-induced muscle regeneration in vivo. In aged muscle, a cell intrinsic defect in MuSC ciliation was associated with the decrease in regenerative capacity. Exogenous activation of Hedgehog signaling, known to be localized in the primary cilium, promoted MuSC expansion, both in vitro and in vivo. Delivery of the small molecule Smoothened agonist (SAG1.3) to muscles of aged mice restored regenerative capacity leading to increased strength post-injury. These findings provide fresh insights into the signaling dysfunction in aged MuSCs and identify the ciliary Hedgehog signaling pathway as a potential therapeutic target to counter the loss of muscle regenerative capacity which accompanies aging.


Subject(s)
Cilia , Muscle, Skeletal , Aging/physiology , Animals , Hedgehog Proteins , Mice , Muscle, Skeletal/physiology , Myoblasts
2.
J Cachexia Sarcopenia Muscle ; 11(5): 1321-1335, 2020 10.
Article in English | MEDLINE | ID: mdl-32677363

ABSTRACT

BACKGROUND: Surgery can lead to significant muscle loss, which increases recovery time and associates with increased mortality. Muscle loss is not uniform, with some patients losing significant muscle mass and others losing relatively little, and is likely to be accompanied by marked changes in circulating metabolites and proteins. Determining these changes may help understand the variability and identify novel therapeutic approaches or markers of muscle wasting. METHODS: To determine the association between muscle loss and circulating metabolites, we studied 20 male patients (median age, 70.5, interquartile range, 62.5-75) undergoing aortic surgery. Muscle mass was determined before and 7 days after surgery and blood samples were taken before surgery, and 1, 3, and 7 days after surgery. The circulating metabolome and proteome were determined using commercial services (Metabolon and SomaLogic). RESULTS: Ten patients lost more than 10% of the cross-sectional area of the rectus femoris (RFCSA ) and were defined as wasting. Metabolomic analysis showed that 557 circulating metabolites were altered following surgery (q < 0.05) in the whole cohort and 104 differed between wasting and non-wasting patients (q < 0.05). Weighted genome co-expression network analysis, identified clusters of metabolites, both before and after surgery, that associated with muscle mass and function (r = -0.72, p = 6 × 10-4 with RFCSA on Day 0, P = 3 × 10-4 with RFCSA on Day 7 and r = -0.73, P = 5 × 10-4 with hand-grip strength on Day 7). These clusters were mainly composed of acyl carnitines and dicarboxylates indicating that pre-existing mitochondrial dysfunction contributes to muscle loss following surgery. Surgery elevated cortisol to the same extent in wasting and non-wasting patients, but the cortisol:cortisone ratio was higher in the wasting patients (Day 3 P = 0.043 and Day 7 P = 0.016). Wasting patients also showed a greater increase in circulating nucleotides 3 days after surgery. Comparison of the metabolome with inflammatory markers identified by SOMAscan® showed that pre-surgical mitochondrial dysfunction was associated with growth differentiation factor 15 (GDF-15) (r = 0.79, P = 2 × 10-4 ) and that GDF-15, interleukin (IL)-8), C-C motif chemokine 23 (CCL-23), and IL-15 receptor subunit alpha (IL-15RA) contributed to metabolic changes in response to surgery. CONCLUSIONS: We show that pre-existing mitochondrial dysfunction and reduced cortisol inactivation contribute to muscle loss following surgery. The data also implicate GDF-15 and IL-15RA in mitochondrial dysfunction.


Subject(s)
Hand Strength , Mitochondria , Ventricular Function, Left , Aged , Humans , Intensive Care Units , Male , Middle Aged , Quadriceps Muscle , Stroke Volume
3.
Physiol Rep ; 7(22): e14289, 2019 11.
Article in English | MEDLINE | ID: mdl-31782241

ABSTRACT

Injured tendons heal through the formation of a fibrovascular scar that has inferior mechanical properties compared to native tendon tissue. Reducing inflammation that occurs as a result of the injury could limit scar formation and improve functional recovery of tendons. Prostaglandin D2 (PGD2 ) plays an important role in promoting inflammation in some injury responses and chronic disease processes, and the inhibition of PGD2 has improved healing and reduced disease burden in animal models and early clinical trials. Based on these findings, we sought to determine the role of PGD2 signaling in the healing of injured tendon tissue. We tested the hypothesis that a potent and specific inhibitor of hematopoietic PGD synthase (HPGDS), GSK2894631A, would improve the recovery of tendons of adult male rats following an acute tenotomy and repair. To test this hypothesis, we performed a full-thickness plantaris tendon tenotomy followed by immediate repair and treated rats twice daily with either 0, 2, or 6 mg/kg of GSK2894631A. Tendons were collected either 7 or 21 days after surgical repair, and mechanical properties of tendons were assessed along with RNA sequencing and histology. While there were some differences in gene expression across groups, the targeted inhibition of HPGDS did not impact the functional repair of tendons after injury, as HPGDS expression was surprisingly low in injured tendons. These results indicate that PGD2 signaling does not appear to be important in modulating the repair of injured tendon tissue.


Subject(s)
Achilles Tendon/injuries , Achilles Tendon/metabolism , Prostaglandin D2/metabolism , Recovery of Function/physiology , Signal Transduction/physiology , Achilles Tendon/drug effects , Animals , Biomechanical Phenomena/drug effects , Biomechanical Phenomena/physiology , Enzyme Inhibitors/pharmacology , Hindlimb/drug effects , Hindlimb/injuries , Hindlimb/metabolism , Male , Prostaglandin D2/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Signal Transduction/drug effects , Tendon Injuries/metabolism
4.
J Physiol ; 597(17): 4615-4625, 2019 09.
Article in English | MEDLINE | ID: mdl-31246276

ABSTRACT

KEY POINTS: Skeletal muscle fatigue limits performance in various physical activities, with exercise intolerance being a key symptom in a broad spectrum of diseases. We investigated whether a small molecule fast skeletal troponin activator (FSTA), CK-2066260, can mitigate muscle fatigue by reducing the cytosolic free [Ca2+ ] required to produce a given submaximal force and hence decreasing the energy requirement. Isolated intact single mouse muscle fibres and rat muscles in-situ treated with CK-2066260 showed improved muscle endurance., which was accompanied by decreased ATP demand and reduced glycogen usage. CK-2066260 treatment improved in-vivo exercise capacity in healthy rats and in a rat model of peripheral artery insufficiency. In conclusion, we show that the FSTA CK-2066260 effectively counteracts muscle fatigue in rodent skeletal muscle in vitro, in situ, and in vivo. This may translate to humans and provide a promising pharmacological treatment to patients suffering from severe muscle weakness and exercise intolerance. ABSTRACT: Skeletal muscle fatigue limits performance during physical exercise and exacerbated muscle fatigue is a prominent symptom among a broad spectrum of diseases. The present study investigated whether skeletal muscle fatigue is affected by the fast skeletal muscle troponin activator (FSTA) CK-2066260, which increases myofibrillar Ca2+ sensitivity and amplifies the submaximal force response. Because more force is produced for a given Ca2+ , we hypothesized that CK-2066260 could mitigate muscle fatigue by reducing the energetic cost of muscle activation. Isolated single mouse muscle fibres were fatigued by 100 repeated 350 ms contractions while measuring force and the cytosolic free [Ca2+ ] or [Mg2+ ] ([Mg2+ ]i ). When starting fatiguing stimulation at matching forces (i.e. lower stimulation frequency with CK-2066260): force was decreased by ∼50% with and by ∼75% without CK-2066260; [Mg2+ ]i was increased by ∼10% with and ∼32% without CK-2066260, reflecting a larger decrease in [ATP] in the latter. The glycogen content in in situ stimulated rat muscles fatigued by repeated contractions at matching forces was about two times higher with than without CK-2066260. Voluntary exercise capacity, assessed by rats performing rotarod exercise and treadmill running, was improved in the presence of CK-2066260. CK-2066260 treatment also increased skeletal muscle fatigue resistance and exercise performance in a rat model of peripheral artery insufficiency. In conclusion, we demonstrate that the FSTA CK-2066260 mitigates skeletal muscle fatigue by reducing the metabolic cost of force generation.


Subject(s)
Muscle Contraction/physiology , Muscle Fatigue/physiology , Muscle Fibers, Fast-Twitch/metabolism , Troponin/metabolism , Animals , Calcium/metabolism , Female , Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , Myofibrils/metabolism , Physical Conditioning, Animal/physiology , Rats , Rats, Sprague-Dawley
5.
Skelet Muscle ; 8(1): 38, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30526662

ABSTRACT

Following publication of the original article [1], the authors flagged that there is a discrepancy with the Availability of data and materials statement on page 12 of the article.

6.
Skelet Muscle ; 8(1): 35, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30424786

ABSTRACT

BACKGROUND: In muscular dystrophy and old age, skeletal muscle repair is compromised leading to fibrosis and fatty tissue accumulation. Therefore, therapies that protect skeletal muscle or enhance repair would be valuable medical treatments. Hypoxia-inducible factors (HIFs) regulate gene transcription under conditions of low oxygen, and HIF target genes EPO and VEGF have been associated with muscle protection and repair. We tested the importance of HIF activation following skeletal muscle injury, in both a murine model and human volunteers, using prolyl hydroxylase inhibitors that stabilize and activate HIF. METHODS: Using a mouse eccentric limb injury model, we characterized the protective effects of prolyl hydroxylase inhibitor, GSK1120360A. We then extended these studies to examine the impact of EPO modulation and infiltrating immune cell populations on muscle protection. Finally, we extended this study with an experimental medicine approach using eccentric arm exercise in untrained volunteers to measure the muscle-protective effects of a clinical prolyl hydroxylase inhibitor, daprodustat. RESULTS: GSK1120360A dramatically prevented functional deficits and histological damage, while accelerating recovery after eccentric limb injury in mice. Surprisingly, this effect was independent of EPO, but required myeloid HIF1α-mediated iNOS activity. Treatment of healthy human volunteers with high-dose daprodustat reduced accumulation of circulating damage markers following eccentric arm exercise, although we did not observe any diminution of functional deficits with compound treatment. CONCLUSION: The results of these experiments highlight a novel skeletal muscle protective effect of prolyl hydroxylase inhibition via HIF-mediated expression of iNOS in macrophages. Partial recapitulation of these findings in healthy volunteers suggests elements of consistent pharmacology compared to responses in mice although there are clear differences between these two systems.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glycine/analogs & derivatives , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Muscle Contraction , Muscle, Skeletal/drug effects , Myalgia/drug therapy , Quinolones/therapeutic use , Adolescent , Adult , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glycine/pharmacology , Glycine/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Myalgia/etiology , Quinolones/pharmacology
7.
Methods Mol Biol ; 1787: 77-86, 2018.
Article in English | MEDLINE | ID: mdl-29736711

ABSTRACT

Adult skeletal muscle contains a population of resident stem cells known as muscle stem cells (MuSC) or satellite cells. This population of cells is required for regeneration of functional myofibers after damage. Aging reduces the proliferative response of satellite cells post-injury. This deficient response is thought to contribute to slowed recovery of muscle function after damage in the elderly and may also contribute to age-related loss of muscle function (sarcopenia). Numerous techniques are now available for the isolation of highly purified satellite cells from mice and humans (Sherwood, et al. Cell 119:543-554, 2004; Cerletti, et al. Cell 134:37-47; 2008; Conboy, et al. Methods Mol Biol 621:165-173, 2010; Bareja, et al. PLoS One 9:e90398; 2014; Castiglioni et al. Stem Cell Rep 2:92-106, 2014; Charville, et al. Stem Cell Rep 5:621-632, 2015; Liu et al. Nat Protoc 10:1612-1624, 2015; Sincennes et al. Methods Mol Biol 1556:41-50, 2017), thus opening an opportunity to use satellite cells in phenotypic screens for regulators of satellite cell proliferation and differentiation. In this chapter, we describe a technique for the prospective isolation of mouse satellite cells that we have recently used in a phenotypic screen of a focused set of small molecules.


Subject(s)
Drug Discovery , Muscle, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Phenotype , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Humans , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism
8.
ACS Med Chem Lett ; 9(4): 354-358, 2018 Apr 12.
Article in English | MEDLINE | ID: mdl-29670700

ABSTRACT

The identification and optimization of the first activators of fast skeletal muscle are reported. Compound 1 was identified from high-throughput screening (HTS) and subsequently found to improve muscle function via interaction with the troponin complex. Optimization of 1 for potency, metabolic stability, and physical properties led to the discovery of tirasemtiv (25), which has been extensively characterized in clinical trials for the treatment of amyotrophic lateral sclerosis.

9.
J Physiol ; 595(5): 1657-1670, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27869319

ABSTRACT

KEY POINTS: We report that the small molecule CK-2066260 selectively slows the off-rate of Ca2+ from fast skeletal muscle troponin, leading to increased myofibrillar Ca2+ sensitivity in fast skeletal muscle. Rodents dosed with CK-2066260 show increased hindlimb muscle force and power in response to submaximal rates of nerve stimulation in situ. CK-2066260 has no effect on free cytosolic [Ca2+ ] during contractions of isolated muscle fibres. We conclude that fast skeletal muscle troponin sensitizers constitute a potential therapy to address an unmet need of improving muscle function in conditions of weakness and premature muscle fatigue. ABSTRACT: Skeletal muscle dysfunction occurs in many diseases and can lead to muscle weakness and premature muscle fatigue. Here we show that the fast skeletal troponin activator, CK-2066260, counteracts muscle weakness by increasing troponin Ca2+ affinity, thereby increasing myofibrillar Ca2+ sensitivity. Exposure to CK-2066260 resulted in a concentration-dependent increase in the Ca2+ sensitivity of ATPase activity in isolated myofibrils and reconstituted hybrid sarcomeres containing fast skeletal muscle troponin C. Stopped-flow experiments revealed a ∼2.7-fold decrease in the Ca2+ off-rate of isolated troponin complexes in the presence of CK-2066260 (6 vs. 17 s-1 under control conditions). Isolated mouse flexor digitorum brevis fibres showed a rapidly developing, reversible and concentration-dependent force increase at submaximal stimulation frequencies. This force increase was not accompanied by any changes in the free cytosolic [Ca2+ ] or its kinetics. CK-2066260 induced a slowing of relaxation, which was markedly larger at 26°C than at 31°C and could be linked to the decreased Ca2+ off-rate of troponin C. Rats dosed with CK-2066260 showed increased hindlimb isometric and isokinetic force in response to submaximal rates of nerve stimulation in situ producing significantly higher absolute forces at low isokinetic velocities, whereas there was no difference in force at the highest velocities. Overall muscle power was increased and the findings are consistent with a lack of effect on crossbridge kinetics. In conclusion, CK-2066260 acts as a fast skeletal troponin activator that may be used to increase muscle force and power in conditions of muscle weakness.


Subject(s)
Calcium/physiology , Imidazoles/pharmacology , Muscle Fibers, Fast-Twitch/drug effects , Myofibrils/drug effects , Pyrazines/pharmacology , Adenosine Triphosphatases/physiology , Animals , Cattle , Female , Hindlimb/drug effects , Hindlimb/physiology , Mice, Inbred C57BL , Muscle Fibers, Fast-Twitch/physiology , Myofibrils/physiology , Rabbits , Rats, Sprague-Dawley , Troponin C/physiology
10.
Aging Cell ; 15(3): 582-4, 2016 06.
Article in English | MEDLINE | ID: mdl-27139744

ABSTRACT

Recent high-profile studies report GDF11 to be a key circulating 'anti-aging' factor. However, a screen of extracellular proteins attempting to identify factors with 'anti-aging' phenotypes in aged murine skeletal muscle satellite cells did not identify GDF11 activity. We have been unable to confirm the reported activity of GDF11, similar to other laboratories offering conflicting data and describe our attempts to do so in this short take.


Subject(s)
Cellular Senescence/drug effects , Growth Differentiation Factors/pharmacology , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Count , HEK293 Cells , Humans , Mice , Recombinant Proteins/pharmacology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism
11.
PLoS One ; 9(5): e96921, 2014.
Article in English | MEDLINE | ID: mdl-24805850

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a motor neuron disease characterized by progressive motor neuron loss resulting in muscle atrophy, declining muscle function, and eventual paralysis. Patients typically die from respiratory failure 3 to 5 years from the onset of symptoms. Tirasemtiv is a fast skeletal troponin activator that sensitizes the sarcomere to calcium; this mechanism of action amplifies the response of muscle to neuromuscular input producing greater force when nerve input is reduced. Here, we demonstrate that a single dose of tirasemtiv significantly increases submaximal isometric force, forelimb grip strength, grid hang time, and rotarod performance in a female transgenic mouse model (B6SJL-SOD1 G93A) of ALS with functional deficits. Additionally, diaphragm force and tidal volume are significantly higher in tirasemtiv-treated female B6SJL-SOD1 G93A mice. These results support the potential of fast skeletal troponin activators to improve muscle function in neuromuscular diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Imidazoles/administration & dosage , Motor Neurons/drug effects , Muscle Strength/drug effects , Pyrazines/administration & dosage , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Motor Neurons/pathology , Muscle Strength/genetics , Muscle, Skeletal/drug effects , Troponin/genetics , Troponin/metabolism
12.
Circ Cardiovasc Genet ; 7(2): 132-143, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24585742

ABSTRACT

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a common genetic disorder caused mainly by mutations in sarcomeric proteins and is characterized by maladaptive myocardial hypertrophy, diastolic heart failure, increased myofilament Ca(2+) sensitivity, and high susceptibility to sudden death. We tested the following hypothesis: correction of the increased myofilament sensitivity can delay or prevent the development of the HCM phenotype. METHODS AND RESULTS: We used an HCM mouse model with an E180G mutation in α-tropomyosin (Tm180) that demonstrates increased myofilament Ca(2+) sensitivity, severe hypertrophy, and diastolic dysfunction. To test our hypothesis, we reduced myofilament Ca(2+) sensitivity in Tm180 mice by generating a double transgenic mouse line. We crossed Tm180 mice with mice expressing a pseudophosphorylated cardiac troponin I (S23D and S24D; TnI-PP). TnI-PP mice demonstrated a reduced myofilament Ca(2+) sensitivity compared with wild-type mice. The development of pathological hypertrophy did not occur in mice expressing both Tm180 and TnI-PP. Left ventricle performance was improved in double transgenic compared with their Tm180 littermates, which express wild-type cardiac troponin I. Hearts of double transgenic mice demonstrated no changes in expression of phospholamban and sarcoplasmic reticulum Ca(2+) ATPase, increased levels of phospholamban and troponin T phosphorylation, and reduced phosphorylation of TnI compared with Tm180 mice. Moreover, expression of TnI-PP in Tm180 hearts inhibited modifications in the activity of extracellular signal-regulated kinase and zinc finger-containing transcription factor GATA in Tm180 hearts. CONCLUSIONS: Our data strongly indicate that reduction of myofilament sensitivity to Ca(2+) and associated correction of abnormal relaxation can delay or prevent development of HCM and should be considered as a therapeutic target for HCM.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Myofibrils/metabolism , Tropomyosin/genetics , Troponin I/genetics , Animals , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Hypertrophic/therapy , Humans , Mice , Mice, Transgenic , Mutation , Phosphorylation , Tropomyosin/metabolism , Troponin I/metabolism , Troponin T/metabolism
13.
PLoS One ; 9(2): e90398, 2014.
Article in English | MEDLINE | ID: mdl-24587351

ABSTRACT

Satellite cells are the chief contributor to skeletal muscle growth and regeneration. The study of mouse satellite cells has accelerated in recent years due to technical advancements in the isolation of these cells. The study of human satellite cells has lagged and thus little is known about how the biology of mouse and human satellite cells compare. We developed a flow cytometry-based method to prospectively isolate human skeletal muscle progenitors from the satellite cell pool using positive and negative selection markers. Results show that this pool is enriched in PAX7 expressing cells that possess robust myogenic potential including the ability to give rise to de novo muscle in vivo. We compared mouse and human satellite cells in culture and identify differences in the elaboration of the myogenic genetic program and in the sensitivity of the cells to cytokine stimulation. These results indicate that not all mechanisms regulating mouse satellite cell activation are conserved in human satellite cells and that such differences may impact the clinical translation of therapeutics validated in mouse models. Thus, the findings of this study are relevant to developing therapies to combat muscle disease.


Subject(s)
Muscle Development/genetics , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Adolescent , Adult , Animals , Biomarkers/metabolism , Female , Flow Cytometry , Gene Expression , Humans , Male , Mice , Middle Aged , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Satellite Cells, Skeletal Muscle/cytology , Species Specificity
14.
Am J Physiol Endocrinol Metab ; 306(2): E150-6, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24253050

ABSTRACT

A splice form of IGF-1, IGF-1Eb, is upregulated after exercise or injury. Physiological responses have been ascribed to the 24-amino acid COOH-terminal peptide that is cleaved from the NH3-terminal 70-amino acid mature IGF-1 protein. This COOH-terminal peptide was termed "mechano-growth factor" (MGF). Activities claimed for the MGF peptide included enhancing muscle satellite cell proliferation and delaying myoblast fusion. As such, MGF could represent a promising strategy to improve muscle regeneration. Thus, at our two pharmaceutical companies, we attempted to reproduce the claimed effect of MGF peptides on human and mouse muscle myoblast proliferation and differentiation in vitro. Concentrations of peptide up to 500 ng/ml failed to increase the proliferation of C2C12 cells or primary human skeletal muscle myoblasts. In contrast, all cell types exhibited a proliferative response to mature IGF-1 or full-length IGF-1Eb. MGF also failed to inhibit the differentiation of myoblasts into myotubes. To address whether the response to MGF was lost in these tissue culture lines, we measured proliferation and differentiation of primary mouse skeletal muscle stem cells exposed to MGF. This, too, failed to demonstrate a significant effect. Finally, we tested whether MGF could alter a separate documented in vitro effect of the peptide, activation of p-ERK, but not p-Akt, in cardiac myocytes. Although a robust response to IGF-1 was observed, there were no demonstrated activating responses from the native or a stabilized MGF peptide. These results call in to question whether there is a physiological role for MGF.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Myoblasts/drug effects , Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/pharmacology , Mice , Myoblasts/physiology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Primary Cell Culture , Protein Processing, Post-Translational , Protein Structure, Tertiary , Stem Cells/physiology
15.
J Muscle Res Cell Motil ; 33(6): 439-48, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22527640

ABSTRACT

Following activation by G-protein-coupled receptor agonists, protein kinase C (PKC) modulates cardiac myocyte function by phosphorylation of intracellular targets including myofilament proteins cardiac troponin I (cTnI) and cardiac myosin binding protein C (cMyBP-C). Since PKC phosphorylation has been shown to decrease myofibril ATPase activity, we hypothesized that PKC phosphorylation of cTnI and cMyBP-C will lower myocyte power output and, in addition, attenuate the elevation in power in response to protein kinase A (PKA)-mediated phosphorylation. We compared isometric force and power generating capacity of rat skinned cardiac myocytes before and after treatment with the catalytic subunit of PKC. PKC increased phosphorylation levels of cMyBP-C and cTnI and decreased both maximal Ca(2+) activated force and Ca(2+) sensitivity of force. Moreover, during submaximal Ca(2+) activations PKC decreased power output by 62 %, which arose from both the fall in force and slower loaded shortening velocities since depressed power persisted even when force levels were matched before and after PKC. In addition, PKC blunted the phosphorylation of cTnI by PKA, reduced PKA-induced spontaneous oscillatory contractions, and diminished PKA-mediated elevations in myocyte power. To test whether altered thin filament function plays an essential role in these contractile changes we investigated the effects of chronic cTnI pseudo-phosphorylation on myofilament function using myocyte preparations from transgenic animals in which either only PKA phosphorylation sites (Ser-23/Ser-24) (PP) or both PKA and PKC phosphorylation sites (Ser-23/Ser-24/Ser-43/Ser-45/T-144) (All-P) were replaced with aspartic acid. Cardiac myocytes from All-P transgenic mice exhibited reductions in maximal force, Ca(2+) sensitivity of force, and power. Similarly diminished power generating capacity was observed in hearts from All-P mice as determined by in situ pressure-volume measurements. These results imply that PKC-mediated phosphorylation of cTnI plays a dominant role in depressing contractility, and, thus, increased PKC isozyme activity may contribute to maladaptive behavior exhibited during the progression to heart failure.


Subject(s)
Cardiac Output/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C/metabolism , Animals , Male , Myocytes, Cardiac/enzymology , Rats , Rats, Sprague-Dawley
16.
Nat Med ; 18(3): 452-5, 2012 Feb 19.
Article in English | MEDLINE | ID: mdl-22344294

ABSTRACT

Limited neural input results in muscle weakness in neuromuscular disease because of a reduction in the density of muscle innervation, the rate of neuromuscular junction activation or the efficiency of synaptic transmission. We developed a small-molecule fast-skeletal-troponin activator, CK-2017357, as a means to increase muscle strength by amplifying the response of muscle when neural input is otherwise diminished secondary to neuromuscular disease. Binding selectively to the fast-skeletal-troponin complex, CK-2017357 slows the rate of calcium release from troponin C and sensitizes muscle to calcium. As a consequence, the force-calcium relationship of muscle fibers shifts leftwards, as does the force-frequency relationship of a nerve-muscle pair, so that CK-2017357 increases the production of muscle force in situ at sub-maximal nerve stimulation rates. Notably, we show that sensitization of the fast-skeletal-troponin complex to calcium improves muscle force and grip strength immediately after administration of single doses of CK-2017357 in a model of the neuromuscular disease myasthenia gravis. Troponin activation may provide a new therapeutic approach to improve physical activity in diseases where neuromuscular function is compromised.


Subject(s)
Calcium/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Diseases/metabolism , Troponin C/agonists , Troponin C/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cattle , Humans , Imidazoles/chemistry , Imidazoles/therapeutic use , Molecular Targeted Therapy , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/pathology , Myasthenia Gravis/drug therapy , Myasthenia Gravis/metabolism , Myasthenia Gravis/pathology , Myosins/isolation & purification , Myosins/metabolism , Neuromuscular Diseases/drug therapy , Neuromuscular Diseases/pathology , Pyrazines/chemistry , Pyrazines/therapeutic use , Rabbits , Rats , Troponin/metabolism , Troponin/physiology
17.
J Mol Cell Cardiol ; 50(3): 442-50, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21047515

ABSTRACT

Familial hypertrophic cardiomyopathy (FHC) is a leading cause of sudden cardiac death among young athletes but the functional effects of the myofilament mutations during FHC-associated ischemia and acidosis, due in part to increased extravascular compressive forces and microvascular dysfunction, are not well characterized. We tested the hypothesis that the FHC-linked tropomyosin (Tm) mutation Tm-E180G alters the contractile response to acidosis via increased myofilament Ca(2+) sensitivity. Intact papillary muscles from transgenic (TG) mice expressing Tm-E180G and exposed to acidic conditions (pH 6.9) exhibited a significantly smaller decrease in normalized isometric tension compared to non-transgenic (NTG) preparations. Times to peak tension and to 90% of twitch force relaxation in TG papillary muscles were significantly prolonged. Intact single ventricular TG myocytes demonstrated significantly less inhibition of unloaded shortening during moderate acidosis (pH 7.1) than NTG myocytes. The peak Ca(2+) transients were not different for TG or NTG at any pH tested. The time constant of re-lengthening was slower in TG myocytes, but not the rate of Ca(2+) decline. TG detergent-extracted fibers demonstrated increased Ca(2+) sensitivity of force and maximal tension compared to NTG at both normal and acidic pH (pH 6.5). Tm phosphorylation was not different between TG and NTG muscles at either pH. Our data indicate that acidic pH diminished developed force in hearts of TG mice less than in NTG due to their inherently increased myofilament Ca(2+) sensitivity, thus potentially contributing to altered energy demands and increased propensity for contractile dysfunction.


Subject(s)
Acidosis/genetics , Acidosis/metabolism , Cardiomyopathy, Hypertrophic, Familial/genetics , Cardiomyopathy, Hypertrophic, Familial/metabolism , Mutation , Tropomyosin/genetics , Acidosis/physiopathology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Calcium/metabolism , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Cells, Cultured , Death, Sudden, Cardiac , Isometric Contraction/physiology , Mice , Mice, Transgenic , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Papillary Muscles/metabolism , Papillary Muscles/physiopathology , Phosphorylation , Tropomyosin/metabolism
18.
Am J Physiol Heart Circ Physiol ; 299(3): H723-30, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20562337

ABSTRACT

ROS, such as H(2)O(2), are a component of pathological conditions in many organ systems and have been reported to be elevated in cardiac pathophysiology. The experiments presented here test the hypothesis that H(2)O(2) induces alterations in cardiac myofilament function by the posttranslational modification of sarcomeric proteins indirectly through PKC signaling. In vitro assessment of actomyosin Mg(2+)-ATPase activity of myofibrillar fractions showed blunted relative ATP consumption in the relaxed state (pCa 8.0) in response to treatment with 0.5 mM H(2)O(2) before myofilament isolation. The effect was attributable to downstream "redox signaling," inasmuch as the direct application of H(2)O(2) to isolated myofibrils did not alter Mg(2+)-ATPase activity. Ca(2+)-ATPase activity, which was used as a measure of myofibrillar myosin function, was unaffected by H(2)O(2). Functional experiments using rat cardiac trabeculae treated with 0.5 or 5 mM H(2)O(2) followed by detergent extraction of membranes demonstrated increased Ca(2+) sensitivity of force production, a faster rate of force redevelopment, and (for 5 mM) decreased maximum tension. Biochemical analysis of myocardial samples treated with 0.5 mM H(2)O(2) demonstrated increased phosphorylation of two sarcomeric proteins: cardiac troponin I and myosin-binding protein-C. These changes were eliminated by a general PKC inhibitor. However, H(2)O(2) and the general PKC activator PMA induced different phosphorylation patterns in cardiomyocytes in which PKC-delta was elevated by viral infection. These data provide evidence that PKC-dependent redox signaling affects the function of cardiac myofilaments and indicate modification of specific proteins through this signaling mechanism.


Subject(s)
Hydrogen Peroxide/metabolism , Myocytes, Cardiac/metabolism , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Sarcomeres/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Analysis of Variance , Animals , Blotting, Western , Ca(2+) Mg(2+)-ATPase/metabolism , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Heart/drug effects , Hydrogen Peroxide/pharmacology , Male , Myocardial Contraction/drug effects , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Sarcomeres/drug effects , Signal Transduction/drug effects
19.
J Biol Chem ; 285(16): 11810-7, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20164197

ABSTRACT

There is evidence for PKC-dependent multisite phosphorylation of cardiac troponin I (cTnI) at Ser-23 and Ser-24 (also PKA sites) in the cardiac-specific N-terminal extension and at Thr-144, a unique residue in the inhibitory region. The functional effect of these phosphorylations in combination is of interest in view of data indicating intramolecular interaction between the N-terminal extension and the inhibitory region of cTnI. To determine the role of PKC-dependent phosphorylation of cTnI on sarcomeric function, we measured contractile regulation at multiple levels of complexity. Ca(2+) binding to thin filaments reconstituted with either cTnI(wild-type) or pseudo-phosphorylated cTnI(S23D/S24D), cTnI(T144E), and cTnI(S23D/S24D/T144E) was determined. Compared with controls regulated by cTnI(wild-type), thin filaments with cTnI(S23D/S24D) and cTnI(S23D/S24D/T144E) exhibited decreased Ca(2+) sensitivity. In contrast, there was no significant difference between Ca(2+) binding to thin filaments with cTnI(wild-type) and with cTnI(T144E). Studies of the pCa-force relations in skinned papillary fibers regulated by these forms of cTnI yielded similar results. However, in both the Ca(2+) binding measurements and the skinned fiber tension measurements, the presence of cTnI(S23D/S24D/T144E) induced a much lower Hill coefficient than either wild type, S23D/S24D, or T144E. These data highlight the importance of thin filament-based cooperative mechanisms in cardiac regulation, with implications for mechanisms of control of function in normal and pathological hearts.


Subject(s)
Protein Kinase C/metabolism , Troponin I/chemistry , Troponin I/metabolism , Amino Acid Substitution , Animals , Binding Sites/genetics , Calcium/metabolism , Cattle , Humans , In Vitro Techniques , Kinetics , Male , Mice , Mutagenesis, Site-Directed , Myocardial Contraction , Myocardium/metabolism , Phosphorylation , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Threonine/chemistry , Troponin I/genetics
20.
J Biol Chem ; 284(8): 5097-106, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19106098

ABSTRACT

There is little direct evidence on the role of myosin regulatory light chain phosphorylation in ejecting hearts. In studies reported here we determined the effects of regulatory light chain (RLC) phosphorylation on in situ cardiac systolic mechanics and in vitro myofibrillar mechanics. We compared data obtained from control nontransgenic mice (NTG) with a transgenic mouse model expressing a cardiac specific nonphosphorylatable RLC (TG-RLC(P-). We also determined whether the depression in RLC phosphorylation affected phosphorylation of other sarcomeric proteins. TG-RLC(P-) demonstrated decreases in base-line load-independent measures of contractility and power and an increase in ejection duration together with a depression in phosphorylation of myosin-binding protein-C (MyBP-C) and troponin I (TnI). Although TG-RLC(P-) displayed a significantly reduced response to beta(1)-adrenergic stimulation, MyBP-C and TnI were phosphorylated to a similar level in TG-RLC(P-) and NTG, suggesting cAMP-dependent protein kinase signaling to these proteins was not disrupted. A major finding was that NTG controls were significantly phosphorylated at RLC serine 15 following beta(1)-adrenergic stimulation, a mechanism prevented in TG-RLC(P-), thus providing a biochemical difference in beta(1)-adrenergic responsiveness at the level of the sarcomere. Our measurements of Ca(2+) tension and Ca(2+)-ATPase rate relations in detergent-extracted fiber bundles from LV trabeculae demonstrated a relative decrease in maximum Ca(2+)-activated tension and tension cost in TG-RLC(P-) fibers, with no change in Ca(2+) sensitivity. Our data indicate that RLC phosphorylation is critical for normal ejection and response to beta(1)-adrenergic stimulation. Our data also indicate that the lack of RLC phosphorylation promotes compensatory changes in MyBP-C and TnI phosphorylation, which when normalized do not restore function.


Subject(s)
Actin Cytoskeleton/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Diseases/metabolism , Myocardial Contraction , Myocardium/metabolism , Myosin Light Chains/metabolism , Actin Cytoskeleton/genetics , Animals , Calcium/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Heart Diseases/genetics , Heart Ventricles/metabolism , Mice , Mice, Transgenic , Myocardial Contraction/genetics , Myosin Light Chains/genetics , Phosphorylation/genetics , Sarcomeres/genetics , Sarcomeres/metabolism , Stroke Volume/genetics , Troponin I/genetics , Troponin I/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...