Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Muscle Res Cell Motil ; 45(2): 79-86, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38367152

ABSTRACT

Neuromuscular junctions are the synapses between motor neurons and skeletal muscle fibers, which mediate voluntary muscle movement. Since neuromuscular junctions are also tightly associated with the capping function of terminal Schwann cells, these synapses have been classically regarded as tripartite chemical synapses. Although evidences from sympathetic innervation of neuromuscular junctions was described approximately a century ago, the essential presence and functional relevance of sympathetic contribution to the maintenance and modulation of neuromuscular junctions was demonstrated only recently. These findings shed light on the pathophysiology of different clinical conditions and can optimize surgical and clinical treatment modalities for skeletal muscle disorders.


Subject(s)
Muscle, Skeletal , Neuromuscular Junction , Sympathetic Nervous System , Neuromuscular Junction/metabolism , Humans , Muscle, Skeletal/innervation , Animals
2.
Int J Mol Sci ; 25(8)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38673723

ABSTRACT

Recent studies have shown that maternal vitamin D deficiency (VDD) causes long-term metabolic changes in offspring. However, little is known about the impact of maternal VDD on offspring endocrine pancreas development and insulin secretion in the adult life of male and female animals. Female rats (Wistar Hannover) were fed either control (1000 IU Vitamin D3/kg), VDD (0 IU Vitamin D3/kg), or a Ca2+-enriched VDD diet (0 IU Vitamin D3/kg + Ca2+ and P/kg) for 6 weeks and during gestation and lactation. At weaning, VDD status was confirmed based on low serum calcidiol levels in dams and pups. Next, male and female offspring were randomly separated and fed a standard diet for up to 90 days. At this age, serum calcidiol levels were restored to normal levels in all groups, but serum insulin levels were decreased in VDD males without affecting glucagon levels, glycemia, or glucose tolerance. Islets isolated from VDD males showed lower insulin secretion in response to different glucose concentrations, but this effect was not observed in VDD females. Furthermore, VDD males, but not females, showed a smaller total pancreatic islet area and lower ß cell mass, an effect that was accompanied by reduced gene expression of Ins1, Ins2, Pdx1, and SLC2A2. The decrease in Pdx1 expression was not related to the methylation profile of the promoter region of this gene. Most of these effects were observed in the male VDD+Ca2+ group, indicating that the effects were not due to alterations in Ca2+ metabolism. These data show that maternal VDD selectively impairs the morphology and function of ß cells in adult male offspring rats and that female offspring are fully protected from these deleterious effects.


Subject(s)
Insulin-Secreting Cells , Insulin , Rats, Wistar , Vitamin D Deficiency , Animals , Female , Insulin-Secreting Cells/metabolism , Male , Vitamin D Deficiency/metabolism , Rats , Pregnancy , Insulin/blood , Insulin/metabolism , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/etiology , Sex Factors , Insulin Secretion
3.
Can J Physiol Pharmacol ; 101(12): 620-629, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37747059

ABSTRACT

The purpose of this study was to characterize the role of ß1-AR signaling and its cross-talk between cardiac renin-angiotensin system and thyroid-hormone-induced cardiac hypertrophy. T3 was administered at 0.5 mg·kg-1·day-1 for 10 days in ß1-KOT3 and WTT3 groups, while control groups received vehicle alone. Echocardiography and myocardial histology was performed; cardiac and serum ANGI/ANGII and ANP and cardiac levels of p-PKA, p-ERK1/2, p-p38-MAPK, p-AKT, p-4EBP1, and ACE were measured. WTT3 showed decreased IVSTd and increased LVEDD versus WTsal (p < 0.05). ß1-KOT3 exhibited lower LVEDD and higher relative IVSTd versus ß1-KOsal, the lowest levels of ejection fraction, and the highest levels of cardiomyocyte diameter (p < 0.05). Cardiac ANP levels decreased in WTT3 versus ß1-KOT3 (p < 0.05). Cardiac ACE expression was increased in T3-treated groups (p < 0.05). Phosphorylated-p38 MAPK levels were higher in WTT3 versus WTsal or ß1-KOT3, p-4EBP1 was elevated in ß1-KO animals, and p-ERK1/2 was up-regulated in ß1-KOT3. These findings suggest that ß1-AR signaling is crucial for TiCH.


Subject(s)
Cardiomyopathy, Restrictive , Mice , Animals , Cardiomyopathy, Restrictive/metabolism , Cardiomyopathy, Restrictive/pathology , Mice, Knockout , Myocardium/metabolism , Thyroid Hormones , Receptors, Adrenergic/metabolism , Angiotensin II/pharmacology
4.
Am J Physiol Heart Circ Physiol ; 323(2): H322-H335, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35714175

ABSTRACT

Clinical data point to adverse cardiovascular events elicited by testosterone replacement therapy. Testosterone is the main hormone used in gender-affirming hormone therapy (GAHT) by transmasculine people. However, the cardiovascular impact of testosterone in experimental models of GAHT remains unknown. Sex hormones modulate T-cell activation, and immune mechanisms contribute to cardiovascular risk. The present study evaluated whether testosterone negatively impacts female cardiovascular function by enhancing Th17 cell-linked effector mechanisms. Female (8 wk old) C57BL/6J mice received testosterone (48 mg/kg/wk) for 8 wk. Male mice were used for phenotypical comparisons. The hormone treatment in female mice increased circulating testosterone to levels observed in male mice. Testosterone increased lean body mass and body mass index, and decreased perigonadal fat mass, mimicking clinical findings. After 8 wk, testosterone decreased endothelium-dependent vasodilation and increased peripheral Th17 cells. After 24 wk, testosterone increased blood pressure in female mice. Ovariectomy did not intensify phenotypical or cardiovascular effects by testosterone. Female mice lacking T and B cells [Rag1 knockout (-/-)], as well as female mice lacking IL-17 receptor (IL-17Ra-/-), did not exhibit vascular dysfunction induced by testosterone. Testosterone impaired endothelium-dependent vasodilation in female mice lacking γδ T cells, similarly to the observed in wild-type female mice. Adoptive transfer of CD4+ T cells restored testosterone-induced vascular dysfunction in Rag1-/- female mice. Together, these data suggest that CD4+ T cells, most likely Th17 cells, are central to vascular dysfunction induced by testosterone in female mice, indicating that changes in immune-cell balance are important in the GAHT in transmasculine people.NEW & NOTEWORTHY Sex hormone-induced cardiovascular events are important undesirable effects in transgender people under GAHT. Studies addressing the cardiovascular impact of GAHT will certainly contribute to improve healthcare services offered to this population. Our study showing that vascular dysfunction, via Th17 cell-related mechanisms, precedes increased blood pressure induced by testosterone in a GAHT mouse model, reveals potential mechanisms involved in GAHT-related cardiovascular events and may provide new markers/targets for clinical practices in transmasculine people.


Subject(s)
Cardiovascular Diseases , Testosterone , Animals , Cardiovascular Diseases/drug therapy , Disease Models, Animal , Female , Gonadal Steroid Hormones , Homeodomain Proteins , Humans , Male , Mice , Mice, Inbred C57BL , Th17 Cells
5.
FASEB J ; 34(9): 12946-12962, 2020 09.
Article in English | MEDLINE | ID: mdl-32772437

ABSTRACT

Although we have shown that catecholamines suppress the activity of the Ubiquitin-Proteasome System (UPS) and atrophy-related genes expression through a cAMP-dependent manner in skeletal muscle from rodents, the underlying mechanisms remain unclear. Here, we report that a single injection of norepinephrine (NE; 1 mg kg-1 ; s.c) attenuated the fasting-induced up-regulation of FoxO-target genes in tibialis anterior (TA) muscles by the stimulation of PKA/CREB and Akt/FoxO1 signaling pathways. In addition, muscle-specific activation of PKA by the overexpression of PKA catalytic subunit (PKAcat) suppressed FoxO reporter activity induced by (1) a wild-type; (2) a non-phosphorylatable; (3) a non-phosphorylatable and non-acetylatable forms of FoxO1 and FoxO3; (4) downregulation of FoxO protein content, and probably by (5) PGC-1α up-regulation. Consistently, the overexpression of the PKAcat inhibitor (PKI) up-regulated FoxO activity and the content of Atrogin-1 and MuRF1, as well as induced muscle fiber atrophy, the latter effect being prevented by the overexpression of a dominant negative (d. n.) form of FoxO (d.n.FoxO). The sustained overexpression of PKAcat induced fiber-type transition toward a smaller, slower, and more oxidative phenotype and improved muscle resistance to fatigue. Taken together, our data provide the first evidence that endogenous PKA activity is required to restrain the basal activity of FoxO and physiologically important to maintain skeletal muscle mass.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Forkhead Box Protein O1/metabolism , Muscle, Skeletal/enzymology , Muscular Atrophy/metabolism , Animals , Cell Line , Forkhead Box Protein O3/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Myoblasts, Skeletal/enzymology , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 113(3): 746-50, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26733679

ABSTRACT

The distribution and function of sympathetic innervation in skeletal muscle have largely remained elusive. Here we demonstrate that sympathetic neurons make close contact with neuromuscular junctions and form a network in skeletal muscle that may functionally couple different targets including blood vessels, motor neurons, and muscle fibers. Direct stimulation of sympathetic neurons led to activation of muscle postsynaptic ß2-adrenoreceptor (ADRB2), cAMP production, and import of the transcriptional coactivator peroxisome proliferator-activated receptor γ-coactivator 1α (PPARGC1A) into myonuclei. Electrophysiological and morphological deficits of neuromuscular junctions upon sympathectomy and in myasthenic mice were rescued by sympathicomimetic treatment. In conclusion, this study identifies the neuromuscular junction as a target of the sympathetic nervous system and shows that sympathetic input is crucial for synapse maintenance and function.


Subject(s)
Health , Homeostasis , Nervous System Diseases/pathology , Neuromuscular Junction/pathology , Sympathetic Nervous System/pathology , Active Transport, Cell Nucleus , Animals , Biosensing Techniques , Cell Nucleus/metabolism , Cyclic AMP/metabolism , Female , Male , Mice, Inbred C57BL , Models, Biological , Muscle, Skeletal/innervation , Neuromuscular Junction/metabolism , Neurons/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Signal Transduction , Sympathectomy , Sympathetic Nervous System/metabolism , Transcription Factors/metabolism
7.
Am J Physiol Heart Circ Physiol ; 313(1): H149-H163, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28526706

ABSTRACT

G protein-coupled receptor kinase isoform 2 (GRK2) has a critical role in physiological and pharmacological responses to endogenous and exogenous substances. Sepsis causes an important cardiovascular dysfunction in which nitric oxide (NO) has a relevant role. The present study aimed to assess the putative effect of inducible NO synthase (NOS2)-derived NO on the activity of GRK2 in the context of septic cardiac dysfunction. C57BL/6 mice were submitted to severe septic injury by cecal ligation and puncture (CLP). Heart function was assessed by isolated and perfused heart, echocardiography, and ß-adrenergic receptor binding. GRK2 was determined by immunofluorescence and Western blot analysis in the heart and isolated cardiac myocytes. Sepsis increased NOS2 expression in the heart, increased plasma nitrite + nitrate levels, and reduced isoproterenol-induced isolated ventricle contraction, whole heart tension development, and ß-adrenergic receptor density. Treatment with 1400W or with GRK2 inhibitor prevented CLP-induced cardiac hyporesponsiveness 12 and 24 h after CLP. Increased labeling of total and phosphorylated GRK2 was detected in hearts after CLP. With treatment of 1400W or in hearts taken from septic NOS2 knockout mice, the activation of GRK2 was reduced. 1400W or GRK2 inhibitor reduced mortality, improved echocardiographic cardiac parameters, and prevented organ damage. Therefore, during sepsis, NOS2-derived NO increases GRK2, which leads to a reduction in ß-adrenergic receptor density, contributing to the heart dysfunction. Isolated cardiac myocyte data indicate that NO acts through the soluble guanylyl cyclase/cGMP/PKG pathway. GRK2 inhibition may be a potential therapeutic target in sepsis-induced cardiac dysfunction.NEW & NOTEWORTHY The main novelty presented here is to show that septic shock induces cardiac hyporesponsiveness to isoproterenol by a mechanism dependent on nitric oxide and mediated by G protein-coupled receptor kinase isoform 2. Therefore, G protein-coupled receptor kinase isoform 2 inhibition may be a potential therapeutic target in sepsis-induced cardiac dysfunction.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Sepsis/metabolism , Animals , Enzyme Activation , Female , G-Protein-Coupled Receptor Kinase 2/genetics , Heart Failure/etiology , Heart Failure/pathology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Sepsis/complications , Signal Transduction
8.
J Bioenerg Biomembr ; 49(5): 399-411, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28918598

ABSTRACT

Intracellular long-chain acyl-CoA synthetases (ACSL) activate fatty acids to produce acyl-CoA, which undergoes ß-oxidation and participates in the synthesis of esterified lipids such as triacylglycerol (TAG). Imbalances in these metabolic routes are closely associated with the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Triacsin C is one of the few compounds that inhibit TAG accumulation into lipid droplets (LD) by suppressing ACSL activity. Here we report that treatment of primary rat hepatocytes with triacsin C at concentrations lower than the IC50 (4.1 µM) for LD formation: (i) diminished LD number in a concentration-dependent manner; (ii) increased mitochondrial amount; (iii) markedly improved mitochondrial metabolism by enhancing the ß-oxidation efficiency, electron transport chain capacity, and degree of coupling - treatment of isolated rat liver mitochondria with the same triacsin C concentrations did not affect the last two parameters; (iv) decreased the GSH/GSSG ratio and elevated the protein carbonyl level, which suggested an increased reactive oxygen species production, as observed in isolated mitochondria. The hepatocyte mitochondrial improvements were not related to either the transcriptional levels of PGC-1α or the content of mTOR and phosphorylated AMPK. Triacsin C at 10 µM induced hepatocyte death by necrosis and/or apoptosis through mechanisms associated with mitochondrial permeability transition pore opening, as demonstrated by experiments using isolated mitochondria. Therefore, triacsin C at sub-IC50 concentrations modulates the lipid imbalance by shifting hepatocytes to a more oxidative state and enhancing the fatty acid consumption, which can in turn accelerate lipid oxidation and reverse NAFLD in long-term therapies.


Subject(s)
Hepatocytes/cytology , Lipid Droplets/drug effects , Triazenes/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Lipid Metabolism/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Organelle Biogenesis , Rats , Triazenes/therapeutic use
9.
An Acad Bras Cienc ; 88(3 Suppl): 1743-1754, 2016.
Article in English | MEDLINE | ID: mdl-27556329

ABSTRACT

Adaptive changes of carbohydrate and lipid metabolism induced by 7, 15, 30, 60, 90, 150 and 200 days of fasting were investigated in red tilapia (Oreochromis sp.). Plasma glucose, lactate and free fatty acids (FFA) levels, liver and muscle glycogen and total lipid contents and rates of FFA release from mesenteric adipose tissue (MAT) were measured. Plasma glucose levels showed significant differences only after 90 days of fasting, when glycemia was 34% lower (50±5mg.dL-1) than fed fish values (74±1mg.dL-1), remaining relatively constant until 200 days of fasting. The content of liver glycogen ("15%) in fed tilapia fell 40% in 7 days of food deprivation. In 60, 90 and 150 days of fasting, plasma FFA levels increased 49%, 64% and 90%, respectively, compared to fed fish values. In agreement with the increase in plasma FFA, fasting induced a clear increase in lipolytic activity of MAT incubated in vitro. Addition of isobutylmethylxanthine (cAMP-phosphodiesterase inhibitor) and isoproterenol (non selective beta adrenergic agonist) to the incubation medium induced a reduction of lipolysis in fasted fish, differently to what was observed in mammal adipose tissue. This study allowed a physiological assessment of red tilapia response to starvation.


Subject(s)
Adipose Tissue/metabolism , Fasting/metabolism , Lipolysis , Tilapia/metabolism , Animals , Tilapia/classification , Time Factors
10.
Amino Acids ; 47(4): 745-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25575490

ABSTRACT

Endurance exercise training as well as leucine supplementation modulates glucose homeostasis and protein turnover in mammals. Here, we analyze whether leucine supplementation alters the effects of endurance exercise on these parameters in healthy mice. Mice were distributed into sedentary (C) and exercise (T) groups. The exercise group performed a 12-week swimming protocol. Half of the C and T mice, designated as the CL and TL groups, were supplemented with leucine (1.5 % dissolved in the drinking water) throughout the experiment. As well known, endurance exercise training reduced body weight and the retroperitoneal fat pad, increased soleus mass, increased VO2max, decreased muscle proteolysis, and ameliorated peripheral insulin sensitivity. Leucine supplementation had no effect on any of these parameters and worsened glucose tolerance in both CL and TL mice. In the soleus muscle of the T group, AS-160(Thr-642) (AKT substrate of 160 kDa) and AMPK(Thr-172) (AMP-Activated Protein Kinase) phosphorylation was increased by exercise in both basal and insulin-stimulated conditions, but it was reduced in TL mice with insulin stimulation compared with the T group. Akt phosphorylation was not affected by exercise but was lower in the CL group compared with the other groups. Leucine supplementation increased mTOR phosphorylation at basal conditions, whereas exercise reduced it in the presence of insulin, despite no alterations in protein synthesis. In trained groups, the total FoxO3a protein content and the mRNA for the specific isoforms E2 and E3 ligases were reduced. In conclusion, leucine supplementation did not potentiate the effects of endurance training on protein turnover, and it also reduced its positive effects on glucose homeostasis.


Subject(s)
Dietary Supplements/analysis , Glucose/metabolism , Leucine/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Female , Homeostasis , Humans , Insulin/metabolism , Mice , Muscle, Skeletal/metabolism , Physical Endurance , Protein Biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Swimming , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
11.
Biochim Biophys Acta ; 1832(10): 1591-604, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23643711

ABSTRACT

The mitochondrial redox state plays a central role in the link between mitochondrial overloading and insulin resistance. However, the mechanism by which the ROS induce insulin resistance in skeletal muscle cells is not completely understood. We examined the association between mitochondrial function and H2O2 production in insulin resistant cells. Our hypothesis is that the low mitochondrial oxygen consumption leads to elevated ROS production by a mechanism associated with reduced PGC1α transcription and low content of phosphorylated CREB. The cells were transfected with either the encoded sequence for catalase overexpression or the specific siRNA for catalase inhibition. After transfection, myotubes were incubated with palmitic acid (500µM) and the insulin response, as well as mitochondrial function and fatty acid metabolism, was determined. The low mitochondrial oxygen consumption led to elevated ROS production by a mechanism associated with ß-oxidation of fatty acids. Rotenone was observed to reduce the ratio of ROS production. The elevated H2O2 production markedly decreased the PGC1α transcription, an effect that was accompanied by a reduced phosphorylation of Akt and CREB. The catalase transfection prevented the reduction in the phosphorylated level of Akt and upregulated the levels of phosphorylated CREB. The mitochondrial function was elevated and H2O2 production reduced, thus increasing the insulin sensitivity. The catalase overexpression improved mitochondrial respiration protecting the cells from fatty acid-induced, insulin resistance. This effect indicates that control of hydrogen peroxide production regulates the mitochondrial respiration preventing the insulin resistance in skeletal muscle cells by a mechanism associated with CREB phosphorylation and ß-oxidation of fatty acids.


Subject(s)
Catalase/metabolism , Hydrogen Peroxide/metabolism , Insulin Resistance , Mitochondria, Muscle/physiology , Animals , Antioxidants/metabolism , Cells, Cultured , Male , Mitochondria, Muscle/enzymology , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Oxygen Consumption , Palmitic Acid/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar
12.
Can J Physiol Pharmacol ; 92(6): 445-54, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24826789

ABSTRACT

The aim of this study was to investigate the changes in the rates of both protein synthesis and breakdown, and the activation of intracellular effectors that control these processes in soleus muscles from growing rats fed a low-protein, high-carbohydrate (LPHC) diet for 15 days. The mass and the protein content, as well as the rate of protein synthesis, were decreased in the soleus from LPHC-fed rats. The availability of amino acids was diminished, since the levels of various essential amino acids were decreased in the plasma of LPHC-fed rats. Overall rate of proteolysis was also decreased, explained by reductions in the mRNA levels of atrogin-1 and MuRF-1, ubiquitin conjugates, proteasome activity, and in the activity of caspase-3. Soleus muscles from LPHC-fed rats showed increased insulin sensitivity, with increased levels of insulin receptor and phosphorylation levels of AKT, which probably explains the inhibition of both the caspase-3 activity and the ubiquitin-proteasome system. The fall of muscle proteolysis seems to represent an adaptive response that contributes to spare proteins in a condition of diminished availability of dietary amino acids. Furthermore, the decreased rate of protein synthesis may be the driving factor to the lower muscle mass gain in growing rats fed the LPHC diet.


Subject(s)
Caspase 3/metabolism , Diet, Protein-Restricted , Dietary Carbohydrates/pharmacology , Muscle, Skeletal/drug effects , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis/drug effects , Proteolysis/drug effects , Ubiquitin/metabolism , Amino Acids/blood , Animals , Cathepsin B/metabolism , Dietary Proteins/administration & dosage , Dietary Proteins/pharmacology , Insulin Resistance , Male , Muscle Proteins/biosynthesis , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, Insulin/metabolism , SKP Cullin F-Box Protein Ligases/biosynthesis , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/biosynthesis
13.
Front Physiol ; 15: 1268380, 2024.
Article in English | MEDLINE | ID: mdl-38318197

ABSTRACT

Resistance exercise (RE) training and pharmacological stimulation of ß2-Adrenoceptors (ß2-ARs) alone can promote muscle hypertrophy and prevent muscle atrophy. Although the activation of the sympathetic nervous system (SNS) is a well-established response during RE, the physiological contribution of the endogenous catecholamines and ß2-ARs to the RE-induced changes on skeletal muscle protein metabolism remains unclear. This study investigated the effects of the ß2-ARs blockade on the acute molecular responses induced by a single bout of RE in rodent skeletal muscles. Male C57BL6/J mice were subjected to a single bout of progressive RE (until exhaustion) on a vertical ladder under ß2-AR blockade with ICI 118,551 (ICI; 10 mg kg-1, i. p.), or vehicle (sterile saline; 0.9%, i. p.), and the gene expression was analyzed in gastrocnemius (GAS) muscles by qPCR. We demonstrated that a single bout of RE acutely increased the circulating levels of stress-associated hormones norepinephrine (NE) and corticosterone (CORT), as well as the muscle phosphorylation levels of AMPK, p38 MAPK and CREB, immediately after the session. The acute increase in the phosphorylation levels of CREB was followed by the upregulation of CREB-target genes Sik1, Ppargc1a and Nr4a3 (a central regulator of the acute RE response), 3 h after the RE session. Conversely, ß2-AR blockade reduced significantly the Sik1 and Nr4a3 mRNA levels in muscles of exercised mice. Furthermore, a single bout of RE stimulated the mRNA levels of the atrophic genes Map1lc3b and Gabarapl1 (autophagy-related genes) and Mstn (a well-known negative regulator of muscle growth). Unexpectedly, the gene expression of Igf-1 or Il-6 were not affected by RE, while the atrophic genes Murf1/Trim63 and Atrogin-1/Mafbx32 (ubiquitin-ligases) were increased only in muscles of exercised mice under ß2-AR blockade. Interestingly, performing a single bout of RE under ß2-AR blockade increased the mRNA levels of Mstn in muscles of exercised mice. These data suggest that ß2-ARs stimulation during acute RE stimulates the hypertrophic gene Nr4a3 and prevents the overexpression of atrophic genes such as Mstn, Murf1/Trim63, and Atrogin-1/Mafbx32 in the first hours of postexercise recovery, indicating that he SNS may be physiologically important to muscle adaptations in response to resistance training.

14.
Metabolism ; 157: 155940, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38878857

ABSTRACT

BACKGROUND AND AIM: Although it is well established that hormones like glucagon stimulates gluconeogenesis via the PKA-mediated phosphorylation of CREB and dephosphorylation of the cAMP-regulated CREB coactivators CRTC2, the role of neural signals in the regulation of gluconeogenesis remains uncertain. METHODS AND RESULTS: Here, we characterize the noradrenergic bundle architecture in mouse liver; we show that the sympathoexcitation induced by acute cold exposure promotes hyperglycemia and upregulation of gluconeogenesis via triggering of the CREB/CRTC2 pathway. Following its induction by dephosphorylation, CRTC2 translocates to the nucleus and drives the transcription of key gluconeogenic genes. Rodents submitted to different models of sympathectomy or knockout of CRTC2 do not activate gluconeogenesis in response to cold. Norepinephrine directly acts in hepatocytes mainly through a Ca2+-dependent pathway that stimulates CREB/CRTC2, leading to activation of the gluconeogenic program. CONCLUSION: Our data demonstrate the importance of the CREB/CRTC2 pathway in mediating effects of hepatic sympathetic inputs on glucose homeostasis, providing new insights into the role of norepinephrine in health and disease.


Subject(s)
Cold Temperature , Cyclic AMP Response Element-Binding Protein , Gluconeogenesis , Liver , Norepinephrine , Transcription Factors , Animals , Gluconeogenesis/physiology , Liver/metabolism , Mice , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Norepinephrine/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Adrenergic Neurons/metabolism , Adrenergic Neurons/physiology , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Hepatocytes/metabolism
15.
Am J Physiol Endocrinol Metab ; 305(12): E1483-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24169047

ABSTRACT

The physiological role of epinephrine in the regulation of skeletal muscle protein metabolism under fasting is unknown. We examined the effects of plasma epinephrine depletion, induced by adrenodemedullation (ADMX), on muscle protein metabolism in fed and 2-day-fasted rats. In fed rats, ADMX for 10 days reduced muscle mass, the cross-sectional area of extensor digitorum longus (EDL) muscle fibers, and the phosphorylation levels of Akt. In addition, ADMX led to a compensatory increase in muscle sympathetic activity, as estimated by the rate of norepinephrine turnover; this increase was accompanied by high rates of muscle protein synthesis. In fasted rats, ADMX exacerbated fasting-induced proteolysis in EDL but did not affect the low rates of protein synthesis. Accordingly, ADMX activated lysosomal proteolysis and further increased the activity of the ubiquitin (Ub)-proteasome system (UPS). Moreover, expression of the atrophy-related Ub ligases atrogin-1 and MuRF1 and the autophagy-related genes LC3b and GABARAPl1 were upregulated in EDL muscles from ADMX-fasted rats compared with sham-fasted rats, and ADMX reduced cAMP levels and increased fasting-induced Akt dephosphorylation. Unlike that observed for EDL muscles, soleus muscle proteolysis and Akt phosphorylation levels were not affected by ADMX. In isolated EDL, epinephrine reduced the basal UPS activity and suppressed overall proteolysis and atrogin-1 and MuRF1 induction following fasting. These data suggest that epinephrine released from the adrenal medulla inhibits fasting-induced protein breakdown in fast-twitch skeletal muscles, and these antiproteolytic effects on the UPS and lysosomal system are apparently mediated through a cAMP-Akt-dependent pathway, which suppresses ubiquitination and autophagy.


Subject(s)
Epinephrine/deficiency , Fasting/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Proteolysis , Adipose Tissue/anatomy & histology , Adipose Tissue/drug effects , Adrenal Medulla/physiology , Adrenal Medulla/surgery , Animals , Body Composition/drug effects , Body Composition/physiology , Catecholamines/blood , Epinephrine/pharmacology , Male , Norepinephrine/blood , Organ Size/drug effects , Rats , Rats, Wistar
16.
Am J Physiol Endocrinol Metab ; 302(1): E123-33, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21952035

ABSTRACT

Although it is well known that administration of the selective ß(2)-adrenergic agonist clenbuterol (CB) protects muscle following denervation (DEN), the underlying molecular mechanism remains unclear. We report that in vivo treatment with CB (3 mg/kg sc) for 3 days induces antiproteolytic effects in normal and denervated rat soleus muscle via distinct mechanisms. In normal soleus muscle, CB treatment stimulates protein synthesis, inhibits Ca(2+)-dependent proteolysis, and increases the levels of calpastatin protein. On the other hand, the administration of CB to DEN rats ameliorates the loss of muscle mass, enhances the rate of protein synthesis, attenuates hyperactivation of proteasomal and lysosomal proteolysis, and suppresses the transcription of the lysosomal protease cathepsin L and of atrogin-1/MAFbx and MuRF1, two ubiquitin (Ub) ligases involved in muscle atrophy. These effects were not associated with alterations in either IGF-I content or Akt phosphorylation levels. In isolated muscles, CB (10(-6) M) treatment significantly attenuated DEN-induced overall proteolysis and upregulation in the mRNA levels of the Ub ligases. Similar responses were observed in denervated muscles exposed to 6-BNZ-cAMP (500 µM), a PKA activator. The in vitro addition of triciribine (10 µM), a selective Akt inhibitor, did not block the inhibitory effects of CB on proteolysis and Ub ligase mRNA levels. These data indicate that short-term treatment with CB mitigates DEN-induced atrophy of the soleus muscle through the stimulation of protein synthesis, downregulation of cathepsin L and Ub ligases, and consequent inhibition of lysosomal and proteasomal activities and that these effects are independent of Akt and possibly mediated by the cAMP/PKA signaling pathway.


Subject(s)
Adrenergic beta-Agonists/therapeutic use , Clenbuterol/therapeutic use , Lysosomes/drug effects , Muscle, Skeletal/drug effects , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Adrenergic beta-Agonists/pharmacology , Animals , Cathepsin L/metabolism , Clenbuterol/pharmacology , Cyclic AMP-Dependent Protein Kinases/chemistry , Enzyme Activators/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , In Vitro Techniques , Lysosomes/enzymology , Male , Muscle Denervation/adverse effects , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/enzymology , Muscular Atrophy/metabolism , Muscular Atrophy/prevention & control , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis/drug effects , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
17.
Mol Metab ; 60: 101492, 2022 06.
Article in English | MEDLINE | ID: mdl-35390501

ABSTRACT

OBJECTIVE: Although it is well established that urocortin 2 (Ucn2), a peptide member of the corticotrophin releasing factor (CRF) family, and its specific corticotrophin-releasing factor 2 receptor (CRF2R) are highly expressed in skeletal muscle, the role of this peptide in the regulation of skeletal muscle mass and protein metabolism remains elusive. METHODS: To elucidate the mechanisms how Ucn2 directly controls protein metabolism in skeletal muscles of normal mice, we carried out genetic tools, physiological and molecular analyses of muscles in vivo and in vitro. RESULTS: Here, we demonstrated that Ucn2 overexpression activated cAMP signaling and promoted an expressive muscle hypertrophy associated with higher rates of protein synthesis and activation of Akt/mTOR and ERK1/2 signaling pathways. Furthermore, Ucn2 induced a decrease in mRNA levels of atrogin-1 and in autophagic flux inferred by an increase in the protein content of LC3-I, LC3-II and p62. Accordingly, Ucn2 reduced both the transcriptional activity of FoxO in vivo and the overall protein degradation in vitro through an inhibition of lysosomal proteolytic activity. In addition, we demonstrated that Ucn2 induced a fast-to-slow fiber type shift and improved fatigue muscle resistance, an effect that was completely blocked in muscles co-transfected with mitogen-activated protein kinase phosphatase 1 (MKP-1), but not with dominant-negative Akt mutant (Aktmt). CONCLUSIONS: These data suggest that Ucn2 triggers an anabolic and anti-catabolic response in skeletal muscle of normal mice probably through the activation of cAMP cascade and participation of Akt and ERK1/2 signaling. These findings open new perspectives in the development of therapeutic strategies to cope with the loss of muscle mass.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Proto-Oncogene Proteins c-akt , Urocortins/metabolism , Adrenocorticotropic Hormone/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Hypertrophy/metabolism , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Mice , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Urocortins/pharmacology
18.
J Cachexia Sarcopenia Muscle ; 13(4): 2175-2187, 2022 08.
Article in English | MEDLINE | ID: mdl-35582969

ABSTRACT

BACKGROUND: Fetal stage is a critical developmental window for the skeletal muscle, but little information is available about the impact of maternal vitamin D (Vit. D) deficiency (VDD) on offspring lean mass development in the adult life of male and female animals. METHODS: Female rats (Wistar Hannover) were fed either a control (1000 IU Vit. D3/kg) or a VDD diet (0 IU Vit. D3/kg) for 6 weeks and during gestation and lactation. At weaning, male and female offspring were randomly separated and received a standard diet up to 180 days old. RESULTS: Vitamin D deficiency induced muscle atrophy in the male (M-VDD) offspring at the end of weaning, an effect that was reverted along the time. Following 180 days, fast-twitch skeletal muscles [extensor digitorum longus (EDL)] from the M-VDD showed a decrease (20%; P < 0.05) in the number of total fibres but an increase in the cross-sectional area of IIB (17%; P < 0.05), IIA (19%; P < 0.05) and IIAX (21%; P < 0.05) fibres. The fibre hypertrophy was associated with the higher protein levels of MyoD (73%; P < 0.05) and myogenin (55% %; P < 0.05) and in the number of satellite cells (128.8 ± 14 vs. 91 ± 7.6 nuclei Pax7 + in the M-CTRL; P < 0.05). M-VDD increased time to fatigue during ex vivo contractions of EDL muscles and showed an increase in the phosphorylation levels of IGF-1/insulin receptor and their downstream targets related to anabolic processes and myogenic activation, including Ser 473 Akt and Ser 21/9 GSK-3ß. In such muscles, maternal VDD induced a compensatory increase in the content of calcitriol (two-fold; P < 0.05) and CYP27B1 (58%; P < 0.05), a metabolizing enzyme that converts calcidiol to calcitriol. Interestingly, most morphological and biochemical changes found in EDL were not observed in slow-twitch skeletal muscles (soleus) from the M-VDD group as well as in both EDL and soleus muscles from the female offspring. CONCLUSIONS: These data show that maternal VDD selectively affects the development of type-II muscle fibres in male offspring rats but not in female offspring rats and suggest that the enhancement of their size and fatigue resistance in fast-twitch skeletal muscle (EDL) is probably due to a compensatory increase in the muscle content of Vit. D in the adult age.


Subject(s)
Muscle Fibers, Slow-Twitch , Vitamin D Deficiency , Animals , Calcitriol/analysis , Calcitriol/metabolism , Calcitriol/pharmacology , Female , Glycogen Synthase Kinase 3 beta/analysis , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/pharmacology , Male , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Vitamin D Deficiency/complications , Vitamin D Deficiency/metabolism
19.
Mol Vis ; 17: 2228-40, 2011.
Article in English | MEDLINE | ID: mdl-21897745

ABSTRACT

PURPOSE: The apoptosis of retinal neurons plays a critical role in the pathogenesis of diabetic retinopathy (DR), but the molecular mechanisms underlying this phenomenon remain unclear. The purpose of this study was to investigate the cellular localization and the expression of microRNA-29b (miR-29b) and its potential target PKR associated protein X (RAX), an activator of the pro-apoptotic RNA-dependent protein kinase (PKR) signaling pathway, in the retina of normal and diabetic rats. METHODS: Retinas were obtained from normal and diabetic rats within 35 days after streptozotocin (STZ) injection. In silico analysis indicated that RAX is a potential target of miR-29b. The cellular localization of miR-29b and RAX was assessed by in situ hybridization and immunofluorescence, respectively. The expression levels of miR-29b and RAX mRNA were evaluated by quantitative reverse transcription PCR (qRT-PCR), and the expression of RAX protein was evaluated by western blot. A luciferase reporter assay and inhibition of endogenous RAX were performed to confirm whether RAX is a direct target of miR-29b as predicted by the in silico analysis. RESULTS: We found that miR-29b and RAX are localized in the retinal ganglion cells (RGCs) and the cells of the inner nuclear layer (INL) of the retinas from normal and diabetic rats. Thus, the expression of miR-29b and RAX, as assessed in the retina by quantitative RT-PCR, reflects their expression in the RGCs and the cells of the INL. We also revealed that RAX protein is upregulated (more than twofold) at 3, 6, 16, and 22 days and downregulated (70%) at 35 days, whereas miR-29b is upregulated (more than threefold) at 28 and 35 days after STZ injection. We did not confirm the computational prediction that RAX is a direct target of miR-29b. CONCLUSIONS: Our results suggest that RAX expression may be indirectly regulated by miR-29b, and the upregulation of this miRNA at the early stage of STZ-induced diabetes may have a protective effect against the apoptosis of RGCs and cells of the INL by the pro-apoptotic RNA-dependent protein kinase (PKR) signaling pathway.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Retinal Ganglion Cells/metabolism , Retinal Horizontal Cells/metabolism , Signal Transduction/genetics , eIF-2 Kinase/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/genetics , Gene Expression Regulation , Genes, Reporter , Homeodomain Proteins/genetics , In Situ Hybridization , Luciferases/analysis , Male , MicroRNAs/genetics , Rats , Rats, Wistar , Retinal Ganglion Cells/cytology , Retinal Horizontal Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , eIF-2 Kinase/genetics
20.
Muscle Nerve ; 44(3): 371-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21996797

ABSTRACT

Phosphodiesterase (PDE) inhibition reduces skeletal muscle atrophy, but the underlying molecular mechanism remains unclear. We used microdialysis to investigate the effects of different PDE inhibitors on interstitial tyrosine concentration as well as proteolytic activity and atrogenes expression in isolated rat muscle. Rolipram, a PDE-4-selective inhibitor, reduced the interstitial tyrosine concentration and rates of muscle protein degradation. The rolipram-induced muscle cAMP increase was accompanied by a decrease in ubiquitin-proteasome system (UPS) activity and atrogin-1 mRNA, a ubiquitin-ligase involved in muscle atrophy. This effect was not associated with Akt phosphorylation but was partially blocked by a protein kinase A inhibitor. Fasting increased atrogin-1, MuRF-1 and LC3b expression, and these effects were markedly suppressed by rolipram. Our data suggest that activation of cAMP signaling by PDE-4 blockade leads to inhibition of UPS activity and atrogenes expression independently of Akt. These findings are important for identifying novel approaches to attenuate muscle atrophy.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Gene Expression/drug effects , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Phosphodiesterase 4 Inhibitors/pharmacology , Proteolysis/drug effects , Rolipram/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Gene Expression/physiology , Male , Microtubule-Associated Proteins/metabolism , Models, Animal , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscular Atrophy/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins , Tyrosine/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL