Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Cell Physiol ; 322(3): C382-C394, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35044855

ABSTRACT

Sarcolipin (SLN) is a small regulatory protein that inhibits the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump. When bound to SERCA, SLN reduces the apparent Ca2+ affinity of SERCA and uncouples SERCA Ca2+ transport from its ATP consumption. As such, SLN plays a direct role in altering skeletal muscle relaxation and energy expenditure. Interestingly, the expression of SLN is dynamic during times of muscle adaptation, in that large increases in SLN content are found in response to development, atrophy, overload, and disease. Several groups have suggested that increases in SLN, especially in dystrophic muscle, are deleterious as it may reduce muscle function and exacerbate already abhorrent intracellular Ca2+ levels. However, there is also significant evidence to show that increased SLN content is a beneficial adaptive mechanism that protects the SERCA pump and activates Ca2+ signaling and adaptive remodeling during times of cell stress. In this review, we first discuss the role for SLN in healthy muscle during both development and overload, where SLN has been shown to activate Ca2+ signaling to promote mitochondrial biogenesis, fiber-type shifts, and muscle hypertrophy. Then, with respect to muscle disease, we summarize the discrepancies in the literature as to whether SLN upregulation is adaptive or maladaptive in nature. This review is the first to offer the concept of SLN hormesis in muscle disease, wherein both too much and too little SLN are detrimental to muscle health. Finally, the underlying mechanisms which activate SLN upregulation are discussed, specifically acknowledging a potential positive feedback loop between SLN and Ca2+ signaling molecules.


Subject(s)
Muscle Development , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Muscular Atrophy/enzymology , Muscular Dystrophies/enzymology , Proteolipids/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Calcium Signaling , Humans , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology
2.
Physiol Rep ; 7(14): e14144, 2019 07.
Article in English | MEDLINE | ID: mdl-31325238

ABSTRACT

Phospholamban (PLN) is an important Ca2+ modulator at the sarcoplasmic reticulum (SR) of striated muscles. It physically interacts and inhibits sarcoplasmic reticulum Ca2+ ATPase (SERCA2) function, whereas a protein kinase A (PKA)-dependent phosphorylation at its serine 16 reverses the inhibition. The underlying mechanism of this post-translational modification, however, remains not fully understood. Using publicly available databases, we identified A-kinase anchoring protein 6 (AKAP6) as a candidate that might play some roles in PLN phosphorylation. Immunofluorescence showed colocalization between GFP-AKAP6 and PLN in transfected HEK-293T cells and cultured mouse neonatal cardiomyocytes (CMNCs). Co-immunoprecipitation confirmed the functional interaction between AKAP6 and PLN in HEK-293T and isolated adult rat cardiomyocytes in response to isoproterenol stimulation. Functionally, AKAP6 promoted Ca2+ uptake activity of SERCA1 in cotransfected HEK-293T cells despite the presence of PLN. These results were further confirmed in adult rat cardiomyocytes. Immunofluorescence showed colocalization of both proteins around the perinuclear region, while protein-protein interaction was corroborated by immunoprecipitation of the nucleus-enriched fraction of rat hearts. Our findings suggest AKAP6 as a novel interacting partner to PLN in HEK-293T and murine cardiomyocytes.


Subject(s)
A Kinase Anchor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Calcium Signaling , Cells, Cultured , HEK293 Cells , Humans , Mice , Protein Binding , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
3.
Behav Brain Res ; 359: 853-860, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30041008

ABSTRACT

Typical responses in muscle following acute aerobic exercise have been well documented, but the responses in brain have remained relatively unexplored. Recent reports suggest that a single bout of aerobic exercise can prime motor regions of the human brain to experience use-dependent plasticity, however, the mechanisms underlying this priming phenomenon are unclear. As a result, we asked whether a graded test to exhaustion (GXT), the most widely employed test to examine relationships between exercise and integrated responses within the musculoskeletal, cardiopulmonary, and neuropsychological systems, would be able to upregulate the expression of plasticity-related proteins in sensorimotor cortex in rats. We examined immediate responses in animals following either a GXT, or two resting conditions: non-exercising treadmill controls (TC), and acclimatization controls (AC). Young, male Sprague-Dawley rats (n = 20) on a reverse light cycle (12 h/12 h) were exposed to a treadmill acclimatization procedure consisting of 8 days of increasing exercise intensity (10 m/min up to 25 m/min) for 10 min at the same time each day. The acclimatization was followed by 2 days of rest to reduce any carryover effects. On testing day, rats performed either a GXT, or rested (TC and AC), were then sacrificed and sensorimotor cortex dissected. Homogenates were probed for a physiological marker of stress (HSP 70), and plasticity-related proteins (CaMKII, GluN2A, GluN1, GluA1, GluA2) by Western blotting analysis. Both our acclimatization protocol and single event GXT yielded no observable differences in protein expression, suggesting that single session exercise does not prime brain via altered plasticity-related protein expression.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Oxygen Consumption/physiology , Physical Conditioning, Animal , Receptors, N-Methyl-D-Aspartate/metabolism , Sensorimotor Cortex/physiology , Analysis of Variance , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Dextrans/metabolism , Exercise Test , Male , Rats , Rats, Sprague-Dawley , Time Factors , Wheat Germ Agglutinin-Horseradish Peroxidase Conjugate/metabolism
4.
Hum Mol Genet ; 27(23): 4094-4102, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30137316

ABSTRACT

Duchenne muscular dystrophy (DMD) is the most severe form of muscular dystrophy affecting 1 in 3500 live male births. Although there is no cure for DMD, therapeutic strategies aimed at enhancing calcineurin signalling and promoting the slow fibre phenotype have shown promise in mdx mice, which is the classical mouse model for DMD. Sarcolipin (SLN) is a small protein that regulates the sarco(endo)plasmic reticulum Ca2+-ATPase pump and its expression is highly upregulated in dystrophic skeletal muscle. We have recently shown that SLN in skeletal muscle amplifies calcineurin signalling thereby increasing myofibre size and the slow fibre phenotype. Therefore, in the present study we sought to determine the physiological impact of genetic Sln deletion in mdx mice, particularly on calcineurin signalling, fibre-type distribution and size and dystrophic pathology. We generated an mdx/Sln-null (mdx/SlnKO) mouse colony and hypothesized that the soleus and diaphragm muscles from these mice would display blunted calcineurin signalling, smaller myofibre sizes, an increased proportion of fast fibres and worsened dystrophic pathology compared with mdx mice. Our results show that calcineurin signalling was impaired in mdx/SlnKO mice as indicated by reductions in utrophin, stabilin-2 and calcineurin expression. In addition, mdx/SlnKO muscles contained smaller myofibres, exhibited a slow-to-fast fibre-type switch that corresponded with reduced expression of mitochondrial proteins and displayed a worsened dystrophic pathology compared with mdx muscles. Altogether, our findings demonstrate a critical role for SLN upregulation in dystrophic muscles and suggest that SLN can be viewed as a potential therapeutic target.


Subject(s)
Calcineurin/genetics , Muscle Proteins/genetics , Muscular Dystrophy, Duchenne/genetics , Proteolipids/genetics , Animals , Cell Adhesion Molecules, Neuronal/genetics , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/physiopathology , Myofibrils/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Signal Transduction , Utrophin/genetics
5.
Biochem Biophys Res Commun ; 495(1): 499-505, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29127005

ABSTRACT

The capacity for skeletal muscle to repair from daily insults as well as larger injuries is a vital component to maintaining muscle health over our lifetime. Given the importance of skeletal muscle for our physical and metabolic well-being, identifying novel factors mediating the growth and repair of skeletal muscle will thus build our foundational knowledge and help lead to potential therapeutic avenues for muscle wasting disorders. To that end, we investigated the expression of T-cell death associated gene 51 (TDAG51) during skeletal muscle repair and studied the response of TDAG51 deficient (TDAG51-/-) mice to chemically-induced muscle damage. TDAG51 mRNA and protein expression within uninjured skeletal muscle is almost undetectable but, in response to chemically-induced muscle damage, protein levels increase by 5 days post-injury and remain elevated for up to 10 days of regeneration. To determine the impact of TDAG51 deletion on skeletal muscle form and function, we compared adult male TDAG51-/- mice with age-matched wild-type (WT) mice. Body and muscle mass were not different between the two groups, however, in situ muscle testing demonstrated a significant reduction in force production both before and after fatiguing contractions in TDAG51-/- mice. During the early phases of the regenerative process (5 days post-injury), TDAG51-/- muscles display a significantly larger area of degenerating muscle tissue concomitant with significantly less regenerating area compared to WT (as demonstrated by embryonic myosin heavy chain expression). Despite these early deficits in regeneration, TDAG51-/- muscles displayed no morphological deficits by 10 days post injury compared to WT mice. Taken together, the data presented herein demonstrate TDAG51 expression to be upregulated in damaged skeletal muscle and its absence attenuates the early phases of muscle regeneration.


Subject(s)
Gene Deletion , Muscle, Skeletal/physiology , Regeneration , Transcription Factors/genetics , Up-Regulation , Animals , Cell Line , Male , Mice, Inbred C57BL , Muscle Fatigue , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , RNA, Messenger/genetics , Transcription Factors/metabolism
6.
Am J Physiol Cell Physiol ; 313(2): C154-C161, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28592414

ABSTRACT

Overexpression of sarcolipin (SLN), a regulator of sarco(endo)plasmic reticulum Ca2+-ATPases (SERCAs), stimulates calcineurin signaling to enhance skeletal muscle oxidative capacity. Some studies have shown that calcineurin may also control skeletal muscle mass and remodeling in response to functional overload and unload stimuli by increasing myofiber size and the proportion of slow fibers. To examine whether SLN might mediate these adaptive responses, we performed soleus and gastrocnemius tenotomy in wild-type (WT) and Sln-null (Sln-/-) mice and examined the overloaded plantaris and unloaded/tenotomized soleus muscles. In the WT overloaded plantaris, we observed ectopic expression of SLN, myofiber hypertrophy, increased fiber number, and a fast-to-slow fiber type shift, which were associated with increased calcineurin signaling (NFAT dephosphorylation and increased stabilin-2 protein content) and reduced SERCA activity. In the WT tenotomized soleus, we observed a 14-fold increase in SLN protein, myofiber atrophy, decreased fiber number, and a slow-to-fast fiber type shift, which were also associated with increased calcineurin signaling and reduced SERCA activity. Genetic deletion of Sln altered these physiological outcomes, with the overloaded plantaris myofibers failing to grow in size and number, and transition towards the slow fiber type, while the unloaded soleus muscles exhibited greater reductions in fiber size and number, and an accelerated slow-to-fast fiber type shift. In both the Sln-/- overloaded and unloaded muscles, these findings were associated with elevated SERCA activity and blunted calcineurin signaling. Thus, SLN plays an important role in adaptive muscle remodeling potentially through calcineurin stimulation, which could have important implications for other muscle diseases and conditions.


Subject(s)
Calcineurin/metabolism , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Proteolipids/genetics , Animals , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/physiology , Muscle, Skeletal/surgery , Proteolipids/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Tenotomy
7.
PLoS One ; 12(3): e0173708, 2017.
Article in English | MEDLINE | ID: mdl-28278204

ABSTRACT

Sarcolipin (SLN) and phospholamban (PLN) are two small proteins that regulate the sarco(endo)plasmic reticulum Ca2+-ATPase pumps. In a recent study, we discovered that Pln overexpression (PlnOE) in slow-twitch type I skeletal muscle fibers drastically impaired SERCA function and caused a centronuclear myopathy-like phenotype, severe muscle atrophy and weakness, and an 8 to 9-fold upregulation of SLN protein in the soleus muscles. Here, we sought to determine the physiological role of SLN upregulation, and based on its role as a SERCA inhibitor, we hypothesized that it would represent a maladaptive response that contributes to the SERCA dysfunction and the overall myopathy observed in the PlnOE mice. To this end, we crossed Sln-null (SlnKO) mice with PlnOE mice to generate a PlnOE/SlnKO mouse colony and assessed SERCA function, CNM pathology, in vitro contractility, muscle mass, calcineurin signaling, daily activity and food intake, and proteolytic enzyme activity. Our results indicate that genetic deletion of Sln did not improve SERCA function nor rescue the CNM phenotype, but did result in exacerbated muscle atrophy and weakness, due to a failure to induce type II fiber compensatory hypertrophy and a reduction in total myofiber count. Mechanistically, our findings suggest that impaired calcineurin activation and resultant decreased expression of stabilin-2, and/or impaired autophagic signaling could be involved. Future studies should examine these possibilities. In conclusion, our study demonstrates the importance of SLN upregulation in combating muscle myopathy in the PlnOE mice, and since SLN is upregulated across several myopathies, our findings may reveal SLN as a novel and universal therapeutic target.


Subject(s)
Calcium-Binding Proteins/physiology , Muscle Fibers, Slow-Twitch/pathology , Muscle Proteins/physiology , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Proteolipids/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/pathology , Animals , Calcium/metabolism , Disease Models, Animal , Female , Ion Transport , Male , Mice , Mice, Knockout , Muscle Contraction , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Sarcoplasmic Reticulum/metabolism , Sequence Deletion
8.
Brain Behav ; 6(6): e00470, 2016 06.
Article in English | MEDLINE | ID: mdl-27134770

ABSTRACT

AIMS: Phospholamban (PLN) and sarcolipin (SLN) are small inhibitory proteins that regulate the sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA) pump. Previous work from our laboratory revealed that in the soleus and gluteus minimus muscles of mice overexpressing PLN (Pln (OE)), SERCA function was impaired, dynamin 2 (3-5 fold) and SLN (7-9 fold) were upregulated, and features of human centronuclear myopathy (CNM) were observed. Here, we performed structural and functional experiments to evaluate whether the diaphragm muscles of the Pln (OE) mouse would exhibit CNM pathology and muscle weakness. METHODS: Diaphragm muscles from Pln (OE) and WT mice were subjected to histological/histochemical/immunofluorescent staining, Ca(2+)-ATPase and Ca(2+) uptake assays, Western blotting, and in vitro electrical stimulation. RESULTS: Our results demonstrate that PLN overexpression reduced SERCA's apparent affinity for Ca(2+) but did not reduce maximal SERCA activity or rates of Ca(2+) uptake. SLN was upregulated 2.5-fold, whereas no changes in dynamin 2 expression were found. With respect to CNM, we did not observe type I fiber predominance, central nuclei, or central aggregation of oxidative activity in diaphragm, although type I fiber hypotrophy was present. Furthermore, in vitro contractility assessment of Pln (OE) diaphragm strips revealed no reductions in force-generating capacity, maximal rates of relaxation or force development, but did indicate that ½ relaxation time was prolonged. CONCLUSIONS: Therefore, the effects of PLN overexpression on skeletal muscle phenotype differ between diaphragm and the postural soleus and gluteus minimus muscles. Our findings here point to differences in SLN expression and type I fiber distribution as potential contributing factors.


Subject(s)
Calcium-Binding Proteins/metabolism , Diaphragm/metabolism , Muscle Contraction/physiology , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/physiopathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Disease Models, Animal , Male , Mice , Muscle Proteins/metabolism , Proteolipids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...