Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Cachexia Sarcopenia Muscle ; 14(5): 2310-2326, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37671684

RESUMO

BACKGROUND: Caveolins are the principal structural components of plasma membrane caveolae. Dominant pathogenic mutations in the muscle-specific caveolin-3 (Cav3) gene isoform, such as the limb girdle muscular dystrophy type 1C (LGMD-1C) P104L mutation, result in dramatic loss of the Cav3 protein and pathophysiological muscle weakness/wasting. We hypothesize that such muscle degeneration may be linked to disturbances in signalling events that impact protein turnover. Herein, we report studies assessing the effects of Cav3 deficiency on mammalian or mechanistic target of rapamycin complex 1 (mTORC1) signalling in skeletal muscle cells. METHODS: L6 myoblasts were stably transfected with Cav3P104L or expression of native Cav3 was abolished by CRISPR/Cas9 genome editing (Cav3 knockout [Cav3KO]) prior to performing subcellular fractionation and immunoblotting, analysis of real-time mitochondrial respiration or fixed cell immunocytochemistry. Skeletal muscle from wild-type and Cav3-/- mice was processed for immunoblot analysis of downstream mTORC1 substrate phosphorylation. RESULTS: Cav3 was detected in lysosomal-enriched membranes isolated from L6 myoblasts and observed by confocal microscopy to co-localize with lysosomal-specific markers. Cav3P104L expression, which results in significant (~95%) loss of native Cav3, or CRISPR/Cas9-mediated Cav3KO, reduced amino acid-dependent mTORC1 activation. The decline in mTORC1-directed signalling was detected by immunoblot analysis of L6 muscle cells and gastrocnemius Cav3-/- mouse muscle as judged by reduced phosphorylation of mTORC1 substrates that play key roles in the initiation of protein synthesis (4EBP1S65 and S6K1T389 ). S6K1T389 and 4EBP1S65 phosphorylation reduced by over 75% and 80% in Cav3KO muscle cells and by over 90% and 30% in Cav3-/- mouse skeletal muscle, respectively. The reduction in protein synthetic capacity in L6 muscle cells was confirmed by analysis of puromycylated peptides using the SUnSET assay. Cav3 loss was also associated with a 26% increase in lysosomal cholesterol, and pharmacological manipulation of lysosomal cholesterol was effective in replicating the reduction in mTORC1 activity observed in Cav3KO cells. Notably, re-expression of Cav3 in Cav3KO myoblasts normalized lysosomal cholesterol content, which coincided with a recovery in protein translation and an associated increase in mTORC1-directed phosphorylation of downstream targets. CONCLUSIONS: Our findings indicate that Cav3 can localize on lysosomal membranes and is a novel regulator of mTORC1 signalling in muscle. Cav3 deficiency associated with the Cav3P104L mutation impairs mTORC1 activation and protein synthetic capacity in skeletal muscle cells, which may be linked to disturbances in lysosomal cholesterol trafficking and contribute to the pathology of LGMD-1C.

2.
Mol Metab ; 68: 101661, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36586434

RESUMO

OBJECTIVE: Previous mechanistic studies on immunometabolism have focused on metabolite-based paradigms of regulation, such as itaconate. Here, we, demonstrate integration of metabolite and kinase-based immunometabolic control by AMP kinase. METHODS: We combined whole cell quantitative proteomics with gene knockout of AMPKα1. RESULTS: Comparing macrophages with AMPKα1 catalytic subunit deletion with wild-type, inflammatory markers are largely unchanged in unstimulated cells, but with an LPS stimulus, AMPKα1 knockout leads to a striking M1 hyperpolarisation. Deletion of AMPKα1 also resulted in increased expression of rate-limiting enzymes involved in itaconate synthesis, metabolism of glucose, arginine, prostaglandins and cholesterol. Consistent with this, we observed functional changes in prostaglandin synthesis and arginine metabolism. Selective AMPKα1 activation also unlocks additional regulation of IL-6 and IL-12 in M1 macrophages. CONCLUSIONS: Together, our results validate AMPK as a pivotal immunometabolic regulator in macrophages.


Assuntos
Proteínas Quinases Ativadas por AMP , Macrófagos , Proteínas Quinases Ativadas por AMP/metabolismo , Macrófagos/metabolismo , Succinatos/metabolismo , Transdução de Sinais/genética
3.
Cell Physiol Biochem ; 54(5): 975-993, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32997464

RESUMO

BACKGROUND/AIMS: Sustained increases in the circulating concentration of saturated fatty acids (SFAs, e.g. palmitate (PA), as seen during obesity, induces a chronic low grade inflammatory state that has been linked to metabolic dysfunction in tissues such as skeletal muscle that is characterized by disturbances in mitochondrial function and heightened production of reactive oxygen species (ROS). In contrast, monounsaturated (MUFAs, e.g. palmitoleate, PO; oleate, OL) and certain polyunsaturated (PUFAs, e.g. linoleate, LO) fatty acids have been shown to protect against some of the harmful metabolic effects induced by SFAs although it currently remains unknown whether this protection is associated with improved morphological and functional changes in mitochondrial biology and redox status in skeletal muscle cells. The aim of the present study was to investigate this issue. METHODS: Rat skeletal (L6) myotubes were subject to sustained 16h incubation with SFAs either alone or in combination with a MUFA (PO, OL) or PUFA (LO) prior to performing subcellular fractionation, immunoblotting, fixed/live cell imaging (for assessment of mitochondrial morphology and ROS) or analysis of real time mitochondrial respiration. RESULTS: Incubation of L6 myotubes with PA or stearate (SFA, C18:0) but not laurate (a medium chain SFA, C12:0) induced a robust increase in proinflammatory NFkB signaling as judged by loss of IkBα and increased expression of IL-6. This heightened SFA-induced proinflammatory tone was associated with increased production of ROS (superoxide and hydrogen peroxide) and significant loss in proteins involved in mitochondrial biogenesis, respiration and morphology (i.e. PGC1α, SDHA, ANT1 and MFN2). Consistent with these changes, PA induced profound fragmentation of the mitochondrial network and a marked reduction in mitochondrial respiratory capacity. These changes were not evident in myotubes incubated with PO, OL or LO alone, and, strikingly, these MUFAs and PUFA not only negated the proinflammatory action of PA, but antagonised the biochemical, morphological and functional changes in mitochondrial biology and ROS production induced in myotubes by the sustained oversupply of PA. CONCLUSION: Our findings indicate that PO, OL and LO exhibit anti-inflammatory and antioxidant characteristics and, significantly, they can ameliorate SFA-induced disturbances in mitochondrial form and function. These observations may have important nutritional implications in developing strategies that could potentially help limit obesity-induced metabolic dysfunction in tissues such as skeletal muscle.


Assuntos
Ácidos Graxos Monoinsaturados/farmacologia , Ácidos Graxos Insaturados/farmacologia , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Animais , Células Cultivadas , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , NF-kappa B/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
4.
J Cachexia Sarcopenia Muscle ; 11(3): 838-858, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32090499

RESUMO

BACKGROUND: Caveolin-3 (Cav3) is the principal structural component of caveolae in skeletal muscle. Dominant pathogenic mutations in the Cav3 gene, such as the Limb Girdle Muscular Dystrophy-1C (LGMD1C) P104L mutation, result in substantial loss of Cav3 and myopathic changes characterized by muscle weakness and wasting. We hypothesize such myopathy may also be associated with disturbances in mitochondrial biology. Herein, we report studies assessing the effects of Cav3 deficiency on mitochondrial form and function in skeletal muscle cells. METHODS: L6 myoblasts were stably transfected with Cav3P104L or expression of native Cav3 repressed by shRNA or CRISPR/Cas9 genome editing prior to performing fixed/live cell imaging of mitochondrial morphology, subcellular fractionation and immunoblotting, or analysis of real time mitochondrial respiration. Skeletal muscle from wild-type and Cav3-/- mice was processed for analysis of mitochondrial proteins by immunoblotting. RESULTS: Caveolin-3 was detected in mitochondrial-enriched membranes isolated from mouse gastrocnemius muscle and L6 myoblasts. Expression of Cav3P104L in L6 myoblasts led to its targeting to the Golgi and loss of native Cav3 (>95%), including that associated with mitochondrial membranes. Cav3P104L reduced mitochondrial mass and induced fragmentation of the mitochondrial network that was associated with significant loss of proteins involved in mitochondrial biogenesis, respiration, morphology, and redox function [i.e. PGC1α, succinate dehyrdogenase (SDHA), ANT1, MFN2, OPA1, and MnSOD). Furthermore, Cav3P104L myoblasts exhibited increased mitochondrial cholesterol and loss of cardiolipin. Consistent with these changes, Cav3P104L expression reduced mitochondrial respiratory capacity and increased myocellular superoxide production. These morphological, biochemical, and functional mitochondrial changes were phenocopied in myoblasts in which Cav3 had been silenced/knocked-out using shRNA or CRISPR. Reduced mitochondrial mass, PGC1α, SDHA, ANT1, and MnSOD were also demonstrable in Cav3-/- mouse gastrocnemius. Strikingly, Cav3 re-expression in Cav3KO myoblasts restored its mitochondrial association and facilitated reformation of a tubular mitochondrial network. Significantly, re-expression also mitigated changes in mitochondrial superoxide, cholesterol, and cardiolipin content and recovered cellular respiratory capacity. CONCLUSIONS: Our results identify Cav3 as an important regulator of mitochondrial homeostasis and reveal that Cav3 deficiency in muscle cells associated with the Cav3P104L mutation invokes major disturbances in mitochondrial respiration and energy status that may contribute to the pathology of LGMD1C.


Assuntos
Caveolina 3/deficiência , Músculo Esquelético/fisiopatologia , Distrofia Muscular do Cíngulo dos Membros/genética , Animais , Humanos , Camundongos , Camundongos Knockout , Distrofia Muscular do Cíngulo dos Membros/patologia , Mutação , Transfecção
5.
Cell Mol Life Sci ; 76(24): 4887-4904, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31101940

RESUMO

Sustained nutrient (fuel) excess, as occurs during obesity and diabetes, has been linked to increased inflammation, impaired mitochondrial homeostasis, lipotoxicity, and insulin resistance in skeletal muscle. Precisely how mitochondrial dysfunction is initiated and whether it contributes to insulin resistance in this tissue remains a poorly resolved issue. Herein, we examine the contribution that an increase in proinflammatory NFkB signalling makes towards regulation of mitochondrial bioenergetics, morphology, and dynamics and its impact upon insulin action in skeletal muscle cells subject to chronic fuel (glucose and palmitate) overloading. We show sustained nutrient excess of L6 myotubes promotes activation of the IKKß-NFkB pathway (as judged by a six-fold increase in IL-6 mRNA expression; an NFkB target gene) and that this was associated with a marked reduction in mitochondrial respiratory capacity (>50%), a three-fold increase in mitochondrial fragmentation and 2.5-fold increase in mitophagy. Under these circumstances, we also noted a reduction in the mRNA and protein abundance of PGC1α and that of key mitochondrial components (SDHA, ANT-1, UCP3, and MFN2) as well as an increase in cellular ROS and impaired insulin action in myotubes. Strikingly, pharmacological or genetic repression of NFkB activity ameliorated disturbances in mitochondrial respiratory function/morphology, attenuated loss of SDHA, ANT-1, UCP3, and MFN2 and mitigated the increase in ROS and the associated reduction in myotube insulin sensitivity. Our findings indicate that sustained oversupply of metabolic fuel to skeletal muscle cells induces heightened NFkB signalling and that this serves as a critical driver for disturbances in mitochondrial function and morphology, redox status, and insulin signalling.


Assuntos
Metabolismo Energético/genética , Inflamação/genética , Mitocôndrias Musculares/metabolismo , NF-kappa B/genética , Nutrientes/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Glucose/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Insulina/metabolismo , Resistência à Insulina/genética , Mitocôndrias Musculares/genética , Mitofagia/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , NF-kappa B/metabolismo , Obesidade/genética , Obesidade/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Transdução de Sinais/genética
6.
Biochim Biophys Acta ; 1857(9): 1403-1411, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27154056

RESUMO

Mitochondrial dysfunction has been associated with obesity-related muscle insulin resistance, but the causality of this association is controversial. The notion that mitochondrial oxidative capacity may be insufficient to deal appropriately with excessive nutrient loads is for example disputed. Effective mitochondrial capacity is indirectly, but largely determined by ATP-consuming processes because skeletal muscle energy metabolism is mostly controlled by ATP demand. Probing the bioenergetics of rat and human myoblasts in real time we show here that the saturated fatty acid palmitate lowers the rate and coupling efficiency of oxidative phosphorylation under conditions it causes insulin resistance. Stearate affects the bioenergetic parameters similarly, whereas oleate and linoleate tend to decrease the rate but not the efficiency of ATP synthesis. Importantly, we reveal that palmitate influences how oxidative ATP supply is used to fuel ATP-consuming processes. Direct measurement of newly made protein demonstrates that palmitate lowers the rate of de novo protein synthesis by more than 30%. The anticipated decrease of energy demand linked to protein synthesis is confirmed by attenuated cycloheximide-sensitivity of mitochondrial respiratory activity used to make ATP. This indirect measure of ATP turnover indicates that palmitate lowers ATP supply reserved for protein synthesis by at least 40%. This decrease is also provoked by stearate, oleate and linoleate, albeit to a lesser extent. Moreover, palmitate lowers ATP supply for sodium pump activity by 60-70% and, in human cells, decreases ATP supply for DNA/RNA synthesis by almost three-quarters. These novel fatty acid effects on energy expenditure inform the 'mitochondrial insufficiency' debate.


Assuntos
Trifosfato de Adenosina/metabolismo , Metabolismo Energético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Ácido Palmítico/farmacologia , Animais , Células Cultivadas , Humanos , Biossíntese de Proteínas/efeitos dos fármacos , Ratos
7.
Biochim Biophys Acta ; 1837(2): 270-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24212054

RESUMO

Insulin is essential for the regulation of fuel metabolism and triggers the uptake of glucose by skeletal muscle. The imported glucose is either stored or broken down, as insulin stimulates glycogenesis and ATP synthesis. The mechanism by which ATP production is increased is incompletely understood at present and, generally, relatively little functional information is available on the effect of insulin on mitochondrial function. In this paper we have exploited extracellular flux technology to investigate insulin effects on the bioenergetics of rat (L6) and human skeletal muscle myoblasts and myotubes. We demonstrate that a 20-min insulin exposure significantly increases (i) the cell respiratory control ratio, (ii) the coupling efficiency of oxidative phosphorylation, and (iii) the glucose sensitivity of anaerobic glycolysis. The improvement of mitochondrial function is explained by an insulin-induced immediate decrease of mitochondrial proton leak. Palmitate exposure annuls the beneficial mitochondrial effects of insulin. Our data improve the mechanistic understanding of insulin-stimulated ATP synthesis, and reveal a hitherto undisclosed insulin sensitivity of cellular bioenergetics that suggests a novel way of detecting insulin responsiveness of cells.


Assuntos
Insulina/farmacologia , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Anaerobiose/efeitos dos fármacos , Animais , Linhagem Celular , Respiração Celular/efeitos dos fármacos , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Músculo Esquelético/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Ácido Palmítico/farmacologia , Prótons , Ratos
8.
FEMS Microbiol Lett ; 328(2): 93-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22171975

RESUMO

Exposure to microorganisms is considered an environmental factor that can contribute to Type 1 diabetes. Insulin-binding proteins (IBPs) on microorganisms may induce production of antibodies that can react with the human insulin receptor (HIR) with possible consequences in developing a diabetic autoimmune response against HIR and insulin. The interaction of insulin with microorganisms was studied by screening 45 microbial species for their ability to bind insulin. Binding assays were performed using labelled insulin to identify insulin-binding components on the microorganisms. Burkholderia multivorans and Burkholderia cenocepacia isolated from patients with cystic fibrosis (CF) and the fish pathogen Aeromonas salmonicida were the only strains of those tested, which showed insulin-binding components on their cell surfaces. Further work with A. salmonicida suggested that the insulin-binding activity of A. salmonicida is due to the A-layer. A mutant of A. salmonicida lacking the A-layer showed binding, but at a much reduced rate suggesting another insulin-binding component in addition to the high affinity of the A-protein. Soluble protein lysates were subjected to Western ligand blotting using peroxidase-labelled insulin to detect IBPs. Two positive IBPs were apparent at approximately 30 and 20 kDa in lysates from Burkholderia strains, but no IBP was detected in A. salmonicida lysates.


Assuntos
Aeromonas salmonicida/química , Proteínas de Bactérias/química , Burkholderia cenocepacia/química , Proteínas de Transporte/química , Insulina/química , Proteínas de Bactérias/isolamento & purificação , Infecções por Burkholderia/microbiologia , Parede Celular/química , Fibrose Cística/microbiologia , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/química , Humanos , Insulina/análogos & derivados
9.
FEMS Immunol Med Microbiol ; 63(2): 217-27, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22077225

RESUMO

The effects on pancreatic ß-cell viability and function of three microbial secondary metabolites pyrrolnitrin, phenazine and patulin were investigated, using the rat clonal pancreatic ß-cell line, INS-1. Cells were exposed to 10-fold serial dilutions (range 0-10 µg mL(-1)) of the purified compounds for 2, 24 and 72 h. After 2 h exposure, only patulin (10 µg mL(-1)) was cytotoxic. All compounds showed significant cytotoxicity after 24 h. None of the compounds altered insulin secretion with 2 and 20 mM glucose after 2 h. However, after 24 h treatment, phenazine and pyrrolnitrin (10 and 100 ng mL(-1)) potentiated insulin production and glucose-stimulated insulin secretion, whereas patulin had no effect. Exposure (24 h) to either phenazine (100 ng mL(-1)) or pyrrolnitrin (10 ng mL(-1)) caused similar increases in the Ca(2+) content of INS-1 cells. The outward membrane current was inhibited after 24 h exposure to either phenazine (100 ng mL(-1)) or pyrrolnitrin (10 or 100 ng mL(-1)). This study presents novel data suggesting that high concentrations of pyrrolnitrin and phenazine are cytotoxic to pancreatic ß-cells and thus possibly diabetogenic, whereas at lower concentrations these agents are nontoxic and may be insulinotropic. The possible role of such agents in the development of cystic fibrosis-related diabetes is discussed.


Assuntos
Bactérias/química , Células Secretoras de Insulina/efeitos dos fármacos , Patulina/toxicidade , Fenazinas/toxicidade , Pirrolnitrina/toxicidade , Animais , Bactérias/metabolismo , Cálcio/análise , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/química , Células Secretoras de Insulina/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Patulina/metabolismo , Fenazinas/metabolismo , Pirrolnitrina/metabolismo , Ratos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA