Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.570
Filter
1.
J Biochem Mol Toxicol ; 38(7): e23750, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38952032

ABSTRACT

The treatment of organophosphate (OP) anticholinesterases currently lacks an effective oxime reactivator of OP-inhibited acetylcholinesterase (AChE) which can penetrate the blood-brain barrier (BBB). Our laboratories have synthesized novel substituted phenoxyalkyl pyridinium oximes and tested them for their ability to promote survival of rats challenged with lethal doses of nerve agent surrogates. These previous studies demonstrated the ability of some of these oximes to promote 24-h survival to rats challenged with a lethal level of highly relevant surrogates for sarin and VX. The reactivation of OP-inhibited AChE in peripheral tissues was likely to be a major contributor to their efficacy in survival of lethal OP challenges. In the present study, twenty of these novel oximes were screened in vitro for reactivation ability for AChE in rat skeletal muscle and serum using two nerve agent surrogates: phthalimidyl isopropyl methylphosphonate (PIMP, a sarin surrogate) and 4-nitrophenyl ethyl methylphosphonate (NEMP, a VX surrogate). The oximes demonstrated a range of 23%-102% reactivation of AChE in vitro across both tissue types. Some of the novel oximes tested in the present study demonstrated the ability to more effectively reactivate AChE in serum than the currently approved oxime, 2-PAM. Therefore, some of these novel oximes have the potential to reverse AChE inhibition in peripheral target tissues and contribute to survival efficacy.


Subject(s)
Acetylcholinesterase , Cholinesterase Inhibitors , Cholinesterase Reactivators , Muscle, Skeletal , Organophosphates , Oximes , Animals , Oximes/pharmacology , Oximes/chemistry , Rats , Acetylcholinesterase/metabolism , Acetylcholinesterase/blood , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/toxicity , Organophosphates/toxicity , Male , Cholinesterase Reactivators/pharmacology , Cholinesterase Reactivators/chemistry , Pyridinium Compounds/pharmacology , Rats, Sprague-Dawley
2.
J Exp Biol ; 227(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39054898

ABSTRACT

Adult, lab-reared, highland deer mice acclimate to hypoxia by increasing reliance on carbohydrates to fuel exercise. Yet neither the underlying mechanisms for this shift in fuel use nor the impact of lifetime hypoxia exposure experienced in high alpine conditions, are fully understood. Thus, we assessed the use of fuel during exercise in wild highland deer mice running in their native environment. We examined a key step in muscle carbohydrate oxidation - the regulation of pyruvate dehydrogenase (PDH) - during exercise at altitude in wild highlanders and in first generation (G1) lab-born and -raised highlanders acclimated to normoxia or hypoxia. PDH activity was also determined in the gastrocnemius of G1 highlanders using an in situ muscle preparation. We found that wild highlanders had a high reliance on carbohydrates while running in their native environment, consistent with data from hypoxia-acclimated G1 highlanders. PDH activity in the gastrocnemius was similar post exercise between G1 and wild highlanders. However, when the gastrocnemius was stimulated at a light work rate in situ, PDH activity was higher in hypoxia-acclimated G1 highlanders and was associated with lower intramuscular lactate levels. These findings were supported by lower PDH kinase 2 protein production in hypoxia-acclimated G1 mice. Our findings indicate that adult phenotypic plasticity in response to low oxygen is sufficient to increase carbohydrate reliance during exercise in highland deer mice. Additionally, variation in PDH regulation with hypoxia acclimation contributes to shifts in whole-animal patterns of fuel use and is likely to improve exercise performance via elevated energy yield per mole of O2. .


Subject(s)
Altitude , Muscle, Skeletal , Peromyscus , Physical Conditioning, Animal , Pyruvate Dehydrogenase Complex , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/enzymology , Peromyscus/physiology , Pyruvate Dehydrogenase Complex/metabolism , Male , Acclimatization , Hypoxia/metabolism , Female
5.
Protein J ; 43(2): 351-361, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38605203

ABSTRACT

It is well known that preslaughter (antemortem) stress such as rough handling, transportation, a negative environment, physical discomfort, lack of consistent routine, and bad feed quality has a big impact on meat quality. The antemortem-induced poor meat quality is characterized by low pH, a pale and exudative appearance, and a soft texture. Previous studies indicate that antemortem stress plays a key role in regulating protein acetylation and glycolysis in postmortem (PM) muscle. However, the underlying molecular and biochemical mechanism is not clearly understood yet. In this study, we investigated the relationship between antemortem and protein acetylation and glycolysis using murine longissimus dorsi muscle isolated from ICR mice and murine muscle cell line C2C12 treated with epinephrine hydrochloride. Because adrenaline secretion increases in stressed animals, epinephrine hydrochloride was intraperitoneally injected epinephrine into mice to simulate pre-slaughter stress in this study to facilitate experimental operations and save experimental costs. Our findings demonstrated that protein acetylation in pyruvate kinase M1 (PKM1) form is significantly reduced by antemortem, and the reduced acetylation subsequently leads to an increase in PKM1 enzymatic activity which causes increased glycolysis in PM muscle. By using molecular approaches, we identified lysine 141 in PKM1 as a critical residue for acetylation. Our results in this study provide useful insight for controlling or improving meat quality in the future.


Subject(s)
Glycolysis , Mice, Inbred ICR , Muscle, Skeletal , Pyruvate Kinase , Animals , Glycolysis/drug effects , Mice , Pyruvate Kinase/metabolism , Acetylation , Muscle, Skeletal/metabolism , Muscle, Skeletal/enzymology , Cell Line , Stress, Physiological , Epinephrine/metabolism
6.
Am J Physiol Cell Physiol ; 326(6): C1669-C1682, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38646781

ABSTRACT

We previously showed that the transaminase inhibitor, aminooxyacetic acid, reduced respiration energized at complex II (succinate dehydrogenase, SDH) in mitochondria isolated from mouse hindlimb muscle. The effect required a reduction in membrane potential with resultant accumulation of oxaloacetate (OAA), a potent inhibitor of SDH. To specifically assess the effect of the mitochondrial transaminase, glutamic oxaloacetic transaminase (GOT2) on complex II respiration, and to determine the effect in intact cells as well as isolated mitochondria, we performed respiratory and metabolic studies in wildtype (WT) and CRISPR-generated GOT2 knockdown (KD) C2C12 myocytes. Intact cell respiration by GOT2KD cells versus WT was reduced by adding carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP) to lower potential. In mitochondria of C2C12 KD cells, respiration at low potential generated by 1 µM FCCP and energized at complex II by 10 mM succinate + 0.5 mM glutamate (but not by complex I substrates) was reduced versus WT mitochondria. Although we could not detect OAA, metabolite data suggested that OAA inhibition of SDH may have contributed to the FCCP effect. C2C12 mitochondria differed from skeletal muscle mitochondria in that the effect of FCCP on complex II respiration was not evident with ADP addition. We also observed that C2C12 cells, unlike skeletal muscle, expressed glutamate dehydrogenase, which competes with GOT2 for glutamate metabolism. In summary, GOT2 KD reduced C2C12 respiration in intact cells at low potential. From differential substrate effects, this occurred largely at complex II. Moreover, C2C12 versus muscle mitochondria differ in complex II sensitivity to ADP and differ markedly in expression of glutamate dehydrogenase.NEW & NOTEWORTHY Impairment of the mitochondrial transaminase, GOT2, reduces complex II (succinate dehydrogenase, SDH)-energized respiration in C2C12 myocytes. This occurs only at low inner membrane potential and is consistent with inhibition of SDH. Incidentally, we observed that C2C12 mitochondria compared with muscle tissue mitochondria differ in sensitivity of complex II respiration to ADP and in the expression of glutamate dehydrogenase.


Subject(s)
Cell Respiration , Membrane Potential, Mitochondrial , Mitochondria, Muscle , Animals , Mice , Aspartate Aminotransferase, Mitochondrial/metabolism , Aspartate Aminotransferase, Mitochondrial/genetics , Cell Differentiation/drug effects , Cell Line , Cell Respiration/drug effects , Electron Transport Complex II/metabolism , Electron Transport Complex II/genetics , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Oxygen Consumption/drug effects , Succinate Dehydrogenase/metabolism , Succinate Dehydrogenase/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism
7.
Biochem J ; 481(9): 601-613, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38592741

ABSTRACT

Knowledge of the primary structure of neuronal NO synthase (nNOS) in skeletal muscle is still conflicting and needs further clarification. To elucidate the expression patterns of nNOS isoforms at both mRNA and protein level, systematic reverse transcription (RT)-PCR and epitope mapping by qualitative immunoblot analysis on skeletal muscle of C57/BL6 mice were performed. The ability of the nNOS isoforms to form aggregates was characterized by native low-temperature polyacrylamide electrophoresis (LT-PAGE). The molecular analysis was focused on the rectus femoris (RF) muscle, a skeletal muscle with a nearly balanced ratio of nNOS α- and ß-isoforms. RT-PCR amplificates from RF muscles showed exclusive exon-1d mRNA expression, either with or without exon-µ. Epitope mapping demonstrated the simultaneous expression of the nNOS splice variants α/µ, α/non-µ, ß/µ and ß/non-µ. Furthermore, immunoblotting suggests that the transition between nNOS α- and ß-isoforms lies within exon-3. In LT-PAGE, three protein nNOS associated aggregates were detected in homogenates of RF muscle and tibialis anterior muscle: a 320 kDa band containing nNOS α-isoforms, while 250 and 300 kDa bands consist of nNOS ß-isoforms that form homodimers or heterodimers with non-nNOS proteins.


Subject(s)
Muscle, Skeletal , Nitric Oxide Synthase Type I , Animals , Male , Mice , Exons , Isoenzymes/metabolism , Isoenzymes/genetics , Mice, Inbred C57BL , Muscle, Skeletal/enzymology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type I/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Food Funct ; 14(7): 3126-3138, 2023 Apr 03.
Article in English | MEDLINE | ID: mdl-36929898

ABSTRACT

The function of skeletal muscles can be markedly hampered by obesity. Ten-eleven translocation 2 (TET2) is an important therapeutic target for ameliorating skeletal muscle dysfunction. Our previous study revealed that punicalagin (PUN) regulated TET2 in obese mice; however, whether PUN can prevent obesity-induced skeletal muscle dysfunction by regulating TET2 remains unclear. In the present study, 40 male C57BL/6J mice were divided into four groups (n = 10 per group): the control (CON) group, the high-fat-diet (HFD, negative control) group, the resveratrol (positive control) group, and the PUN group. The ratio of gastrocnemius weight to body weight (0.0097 ± 0.0016 vs. 0.0080 ± 0.0011), the grip strength (120.04 g ± 11.10 vs. 98.89 g ± 2.79), and the muscle fiber count (314.56 per visual field ± 92.73 vs. 236.44 per visual field ± 50.58) in the PUN group were higher than those in the HFD group. Moreover, the levels of the TET2 protein, 5-hydroxymethylcytosine (5hmC), and 5-formylcytosine (5fC) in skeletal muscles were significantly lower in the HFD group than those in the CON group; these levels increased after PUN treatment. Compared with the HFD group, the phosphorylation level of AMP-activated protein kinase (AMPK) α in the PUN group was higher, which effectively enhanced the stability of the TET2 protein. Besides, the ratio of (succinic acid + fumaric acid)/α-ketoglutarate in the PUN group was lower than that in the HFD group (43.21 ± 12.42 vs. 99.19 ± 37.07), and a lower ratio led to a higher demethylase activity of TET2 in the PUN group than in the HFD group. This study highlights that PUN supplementation protects against obesity-induced impairment of the skeletal muscle function via regulating the protein stability of TET2 and the enzymatic activity of TET2 demethylation.


Subject(s)
DNA-Binding Proteins , Dioxygenases , Hydrolyzable Tannins , Muscle, Skeletal , Obesity , Hydrolyzable Tannins/administration & dosage , Hydrolyzable Tannins/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiopathology , Diet, High-Fat/adverse effects , Obesity/complications , Obesity/physiopathology , Obesity/therapy , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Dioxygenases/metabolism , Male , Animals , Mice , Mice, Inbred C57BL , Body Weight/drug effects , AMP-Activated Protein Kinases/metabolism
9.
Science ; 379(6628): 185-190, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36634192

ABSTRACT

Hummingbirds possess distinct metabolic adaptations to fuel their energy-demanding hovering flight, but the underlying genomic changes are largely unknown. Here, we generated a chromosome-level genome assembly of the long-tailed hermit and screened for genes that have been specifically inactivated in the ancestral hummingbird lineage. We discovered that FBP2 (fructose-bisphosphatase 2), which encodes a gluconeogenic muscle enzyme, was lost during a time period when hovering flight evolved. We show that FBP2 knockdown in an avian muscle cell line up-regulates glycolysis and enhances mitochondrial respiration, coincident with an increased mitochondria number. Furthermore, genes involved in mitochondrial respiration and organization have up-regulated expression in hummingbird flight muscle. Together, these results suggest that FBP2 loss was likely a key step in the evolution of metabolic muscle adaptations required for true hovering flight.


Subject(s)
Adaptation, Physiological , Birds , Flight, Animal , Fructose-Bisphosphatase , Gluconeogenesis , Muscle, Skeletal , Animals , Birds/genetics , Birds/metabolism , Energy Metabolism/genetics , Flight, Animal/physiology , Gluconeogenesis/genetics , Adaptation, Physiological/genetics , Fructose-Bisphosphatase/genetics , Muscle, Skeletal/enzymology
10.
Muscle Nerve ; 67(2): 169-176, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36420650

ABSTRACT

INTRODUCTION/AIMS: Rapid-stretch nerve injuries represent a substantial treatment challenge. No study has examined motor neuron connection after rapid-stretch injury. Our objective in this study was to characterize the electrophysiological properties of graded rapid-stretch nerve injury and assess motor neuron health using retrograde labeling and muscle adenosine triphosphatase (ATPase) histology. METHODS: Male C57BL/6 mice (n = 6 per group) were rapid-stretch injured at four levels of severity: sham injury, stretch within elastic modulus, inelastic deformation, and stretch rupture. Serial compound muscle action potential (CMAP) and motor unit number estimation (MUNE) measurements were made for 48 days, followed by retrograde labeling and muscle ATPase histology. RESULTS: Elastic injuries showed no durable abnormalities. Inelastic injury demonstrated profound initial reduction in CMAP and MUNE (P < .036) on day 2, with partial recovery by day 14 after injury (CMAP: 40% baseline, P = .003; MUNE: 55% baseline, P = .033). However, at the experimental endpoint, CMAP had recovered to baseline with only limited improvement in MUNE. Inelastic injury led to reduced retrograde-labeled neurons and grouped fiber type histology. Rupture injury had severe and nonrecovering electrophysiological impairment, dramatically reducing labeled neurons (P = .005), and atrophic or type 1 muscle fibers. There was an excellent correlation between MUNE and retrograde-labeled tibial motor neurons across injury severities (R2  = 0.96). DISCUSSION: There was no significant electrophysiological derangement in low-severity injuries but there was recoverable conduction block in inelastic injury with slow recovery, potentially due to collateral sprouting. Rupture injuries yielded permanent failure of injured axons to reinnervate. These results provide insight into the pathophysiology of clinical injuries and recovery.


Subject(s)
Peripheral Nerve Injuries , Rupture , Animals , Male , Mice , Action Potentials/physiology , Adenosine Triphosphatases/analysis , Mice, Inbred C57BL , Muscle, Skeletal/enzymology , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Elastic Modulus , Rupture/physiopathology , Peripheral Nerve Injuries/physiopathology , Motor Neurons/pathology
11.
Front Endocrinol (Lausanne) ; 13: 957182, 2022.
Article in English | MEDLINE | ID: mdl-36072929

ABSTRACT

The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) pump is responsible for the transport of Ca2+ from the cytosol into the sarcoplasmic reticulum at the expense of ATP, making it a regulator of both muscle relaxation and muscle-based energy expenditure. Neurogranin (Ng) is a small protein that negatively regulates calcineurin signaling. Calcineurin is Ca2+/calmodulin dependent phosphatase that promotes the oxidative fibre type in skeletal muscle and regulates muscle-based energy expenditure. A recent study has shown that calcineurin activation reduces SERCA Ca2+ transport efficiency, ultimately raising energy expenditure. Since the biomedical view of obesity states that it arises as an imbalance between energy intake and expenditure which favors the former, we questioned whether heterozygous Ng deletion (Ng+/- ) would reduce SERCA efficiency and increase energy expenditure in female mice fed a high-fat diet (HFD). Young (3-4-month-old) female wild type (WT) and Ng+/- mice were fed a HFD for 12 weeks with their metabolic profile being analyzed using metabolic cages and DXA scanning, while soleus SERCA efficiency was measured using SERCA specific Ca2+ uptake and ATPase activity assays. Ng+/- mice showed significantly less cage ambulation compared to WT mice but this did not lead to any added weight gain nor changes in daily energy expenditure, glucose or insulin tolerance despite a similar level of food intake. Furthermore, we observed significant reductions in SERCA's apparent coupling ratio which were associated with significant reductions in SERCA1 and phospholamban content. Thus, our results show that Ng regulates SERCA pump efficiency, and future studies should further investigate the potential cellular mechanisms.


Subject(s)
Muscle, Skeletal , Neurogranin , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animals , Calcineurin/metabolism , Diet, High-Fat/adverse effects , Female , Gene Knockdown Techniques , Mice , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Neurogranin/genetics , Neurogranin/metabolism , Proteolipids/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
12.
Int J Mol Sci ; 23(13)2022 Jul 04.
Article in English | MEDLINE | ID: mdl-35806430

ABSTRACT

Glucose 6-P dehydrogenase (G6PD) is the first rate-limiting enzyme in pentose phosphate pathway (PPP), and it is proverbial that G6PD is absent in skeletal muscle. However, how and why G6PD is down-regulated during skeletal muscle development is unclear. In this study, we confirmed the expression of G6PD was down-regulated during myogenesis in vitro and in vivo. G6PD was absolutely silent in adult skeletal muscle. Histone H3 acetylation and DNA methylation act together on the expression of G6PD. Neither knock-down of G6PD nor over-expression of G6PD affects myogenic differentiation. Knock-down of G6PD significantly promotes the sensitivity and response of skeletal muscle cells to insulin; over-expression of G6PD significantly injures the sensitivity and response of skeletal muscle cells to insulin. High-fat diet treatment impairs insulin signaling by up-regulating G6PD, and knock-down of G6PD rescues the impaired insulin signaling and glucose uptake caused by high-fat diet treatment. Taken together, this study explored the importance of G6PD deficiency during myogenic differentiation, which provides new sight to treat insulin resistance and type-2 diabetes.


Subject(s)
Glucosephosphate Dehydrogenase Deficiency , Glucosephosphate Dehydrogenase , Insulin , Muscle, Skeletal , Adult , Glucose/metabolism , Glucose 1-Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase Deficiency/genetics , Glucosephosphate Dehydrogenase Deficiency/metabolism , Humans , Insulin/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism
13.
Int J Mol Sci ; 23(3)2022 Feb 03.
Article in English | MEDLINE | ID: mdl-35163674

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that shows progressive muscle weakness. A few treatments exist including symptomatic therapies, which can prolong survival or reduce a symptom; however, no fundamental therapies have been found. As a therapeutic strategy, enhancing muscle force is important for patients' quality of life. In this study, we focused on skeletal muscle-specific myosin regulatory light chain kinase (skMLCK), which potentially enhances muscle contraction, as overexpression of skMLCK was thought to improve muscle function. The adeno-associated virus serotype 6 encoding skMLCK (AAV6/skMLCK) and eGFP (control) was produced and injected intramuscularly into the lower limbs of SOD1G37R mice, which are a familial ALS model. AAV6/skMLCK showed the successful expression of skMLCK in the muscle tissues. Although the control did not affect the muscle force in both of the WT and SOD1G37R mice, AAV6/skMLCK enhanced the twitch force of SOD1G37R mice and the tetanic force of WT and SOD1G37R mice. These results indicate that overexpression of skMLCK can enhance the tetanic force of healthy muscle as well as rescue weakened muscle function. In conclusion, the gene transfer of skMLCK has the potential to be a new therapy for ALS as well as for other neuromuscular diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Dependovirus/metabolism , Gene Transfer Techniques , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiopathology , Myosin-Light-Chain Kinase/genetics , Animals , Biomechanical Phenomena , Disease Models, Animal , Genetic Vectors/metabolism , HEK293 Cells , Humans , Injections, Intramuscular , Mice, Inbred C57BL , Tetany
14.
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
15.
Can J Physiol Pharmacol ; 100(4): 361-370, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34695364

ABSTRACT

Sarco(endo)plasmic reticulum calcium (Ca2+) ATPase (SERCA) transports Ca2+ in muscle. Impaired SERCA activity may contribute to diabetic myopathy. Sirtuin (SIRT) 3 regulates muscle metabolism and function; however, it is unknown if SIRT3 regulates muscle SERCA activity or acetylation. We determined if SIRT3 overexpression enhances SERCA activity in mouse gastrocnemius muscle and if SIRT3 overexpression preserves gastrocnemius SERCA activity in a model of type 2 diabetes, induced by high fat - high sucrose (HFHS) feeding. We also determined if the acetylation status of SERCA proteins in mouse gastrocnemius is altered by SIRT3 overexpression or HFHS feeding. Wild-type (WT) and SIRT3 transgenic (SIRT3TG) mice, overexpressing SIRT3 in skeletal muscle, were fed a standard or HFHS diet for 4 months. SIRT3TG and WT mice developed obesity and glucose intolerance after 4 months of HFHS feeding. SERCA Vmax was higher in gastrocnemius of SIRT3TG mice compared with WT mice. HFHS-fed mice had lower SERCA1a protein levels and lower SERCA Vmax in their gastrocnemius than control-fed mice. The decrease in SERCA Vmax in gastrocnemius muscle due to HFHS feeding was attenuated by SIRT3 overexpression in HFHS-fed SIRT3TG mice. SERCA1a and SERCA2a acetylation in mouse gastrocnemius was not altered by genotype or diet. These findings suggest SIRT3 overexpression improves SERCA function in mouse skeletal muscle.


Subject(s)
Diabetes Mellitus, Type 2 , Muscle, Skeletal/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sirtuin 3 , Animals , Calcium/metabolism , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress , Mice , Sarcoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sirtuin 3/genetics , Sirtuin 3/metabolism , Sucrose/metabolism
16.
Cells ; 10(11)2021 11 02.
Article in English | MEDLINE | ID: mdl-34831205

ABSTRACT

Rho guanosine triphosphate hydrolases (GTPases) are molecular switches that cycle between an inactive guanosine diphosphate (GDP)-bound and an active guanosine triphosphate (GTP)-bound state during signal transduction. As such, they regulate a wide range of both cellular and physiological processes. In this review, we will summarize recent work on the role of Rho GTPase-regulated pathways in skeletal muscle development, regeneration, tissue mass homeostatic balance, and metabolism. In addition, we will present current evidence that links the dysregulation of these GTPases with diseases caused by skeletal muscle dysfunction. Overall, this information underscores the critical role of a number of members of the Rho GTPase subfamily in muscle development and the overall metabolic balance of mammalian species.


Subject(s)
Homeostasis , Muscle Development , Muscle, Skeletal/enzymology , Muscle, Skeletal/growth & development , rho GTP-Binding Proteins/metabolism , Animals , Humans , Muscular Diseases/enzymology , Muscular Diseases/pathology , Regeneration/physiology
17.
Cells ; 10(11)2021 11 05.
Article in English | MEDLINE | ID: mdl-34831250

ABSTRACT

Skeletal muscle regeneration is a complex process involving crosstalk between immune cells and myogenic precursor cells, i.e., satellite cells. In this scenario, macrophage recruitment in damaged muscles is a mandatory step for tissue repair since pro-inflammatory M1 macrophages promote the activation of satellite cells, stimulating their proliferation and then, after switching into anti-inflammatory M2 macrophages, they prompt satellite cells' differentiation into myotubes and resolve inflammation. Here, we show that acid sphingomyelinase (ASMase), a key enzyme in sphingolipid metabolism, is activated after skeletal muscle injury induced in vivo by the injection of cardiotoxin. ASMase ablation shortens the early phases of skeletal muscle regeneration without affecting satellite cell behavior. Of interest, ASMase regulates the balance between M1 and M2 macrophages in the injured muscles so that the absence of the enzyme reduces inflammation. The analysis of macrophage populations indicates that these events depend on the altered polarization of M1 macrophages towards an M2 phenotype. Our results unravel a novel role of ASMase in regulating immune response during muscle regeneration/repair and suggest ASMase as a supplemental therapeutic target in conditions of redundant inflammation that impairs muscle recovery.


Subject(s)
Macrophages/metabolism , Macrophages/pathology , Muscle, Skeletal/physiology , Regeneration/physiology , Sphingomyelin Phosphodiesterase/metabolism , Animals , Cell Differentiation , Cell Polarity , Cell Proliferation , Enzyme Activation , Inflammation/pathology , Mice, Knockout , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Phenotype , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction , Sphingomyelin Phosphodiesterase/deficiency
18.
Cells ; 10(11)2021 11 09.
Article in English | MEDLINE | ID: mdl-34831312

ABSTRACT

Skeletal muscle regeneration is triggered by local inflammation and is accompanied by phagocytosis of dead cells at the injury site. Efferocytosis regulates the inflammatory program in macrophages by initiating the conversion of their inflammatory phenotype into the healing one. While pro-inflammatory cytokines induce satellite cell proliferation and differentiation into myoblasts, growth factors, such as GDF3, released by healing macrophages drive myoblast fusion and myotube growth. Therefore, improper efferocytosis may lead to impaired muscle regeneration. Transglutaminase 2 (TG2) is a versatile enzyme participating in efferocytosis. Here, we show that TG2 ablation did not alter the skeletal muscle weights or sizes but led to the generation of small size myofibers and to decreased grip force in TG2 null mice. Following cardiotoxin-induced injury, the size of regenerating fibers was smaller, and the myoblast fusion was delayed in the tibialis anterior muscle of TG2 null mice. Loss of TG2 did not affect the efferocytic capacity of muscle macrophages but delayed their conversion to Ly6C-CD206+, GDF3 expressing cells. Finally, TG2 promoted myoblast fusion in differentiating C2C12 myoblasts. These results indicate that TG2 expressed by both macrophages and myoblasts contributes to proper myoblast fusion, and its ablation leads to impaired muscle development and regeneration in mice.


Subject(s)
Muscle, Skeletal/enzymology , Muscle, Skeletal/physiology , Protein Glutamine gamma Glutamyltransferase 2/deficiency , Regeneration , Animals , Biomechanical Phenomena , Cell Differentiation , Cell Fusion , Cell Line , Cell Proliferation , Collagen/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle Development/genetics , Muscle Fatigue , Myoblasts/metabolism , Necrosis , Neutrophils/metabolism , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Satellite Cells, Skeletal Muscle/pathology , Time Factors
19.
Int J Mol Med ; 48(5)2021 11.
Article in English | MEDLINE | ID: mdl-34549305

ABSTRACT

Calpains are a family of Ca2+­dependent cysteine proteases that participate in various cellular processes. Calpain 3 (CAPN3) is a classical calpain with unique N­terminus and insertion sequence 1 and 2 domains that confer characteristics such as rapid autolysis, Ca2+­independent activation and Na+ activation of the protease. CAPN3 is the only muscle­specific calpain that has important roles in the promotion of calcium release from skeletal muscle fibers, calcium uptake of sarcoplasmic reticulum, muscle formation and muscle remodeling. Studies have indicated that recessive mutations in CAPN3 cause limb­girdle muscular dystrophy (MD) type 2A and other types of MD; eosinophilic myositis, melanoma and epilepsy are also closely related to CAPN3. In the present review, the characteristics of CAPN3, its biological functions and roles in the pathogenesis of a number of disorders are discussed.


Subject(s)
Calpain/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Diseases/enzymology , Muscular Diseases/pathology , Animals , Calpain/chemistry , Enzyme Activation , Humans , Models, Biological , Organ Specificity
20.
Biomed Pharmacother ; 143: 112188, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34563947

ABSTRACT

An extract from Artemisia dracunculus L. (termed PMI-5011) improves glucose homeostasis by enhancing insulin action and reducing ectopic lipid accumulation, while increasing fat oxidation in skeletal muscle tissue in obese insulin resistant male mice. A chalcone, DMC-2, in PMI-5011 is the major bioactive that enhances insulin signaling and activation of AKT. However, the mechanism by which PMI-5011 improves lipid metabolism is unknown. AMPK is the cellular energy and metabolic sensor and a key regulator of lipid metabolism in muscle. This study examined PMI-5011 activation of AMPK signaling using murine C2C12 muscle cell culture and skeletal muscle tissue. Findings show that PMI-5011 increases Thr172-phosphorylation of AMPK in muscle cells and skeletal muscle tissue, while hepatic AMPK activation by PMI-5011 was not observed. Increased AMPK activity by PMI-5011 affects downstream signaling of AMPK, resulting in inhibition of ACC and increased SIRT1 protein levels. Selective deletion of DMC-2 from PMI-5011 demonstrates that compounds other than DMC-2 in a "DMC-2 knock out extract" (KOE) are responsible for AMPK activation and its downstream effects. Compared to 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) and metformin, the phytochemical mixture characterizing the KOE appears to more efficiently activate AMPK in muscle cells. KOE-mediated AMPK activation was LKB-1 independent, suggesting KOE does not activate AMPK via LKB-1 stimulation. Through AMPK activation, compounds in PMI-5011 may regulate lipid metabolism in skeletal muscle. Thus, the AMPK-activating potential of the KOE adds therapeutic value to PMI-5011 and its constituents in treating insulin resistance or type 2 diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Artemisia , Enzyme Activators/pharmacology , Hypoglycemic Agents/pharmacology , Insulin Resistance , Muscle, Skeletal/drug effects , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Artemisia/chemistry , Cell Line , Diet, High-Fat , Disease Models, Animal , Enzyme Activation , Enzyme Activators/isolation & purification , Hypoglycemic Agents/isolation & purification , Male , Metformin/pharmacology , Mice, Inbred C57BL , Muscle, Skeletal/enzymology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/enzymology , Phosphorylation , Phytochemicals/isolation & purification , Plant Extracts/isolation & purification , Ribonucleotides/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL