Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 1604, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452345

ABSTRACT

Myostatin is a negative regulator of muscle mass and its inhibition represents a promising strategy for the treatment of muscle disorders and type 2 diabetes. However, there is currently no clinically effective myostatin inhibitor, and therefore novel methods are required. We evaluated the use of antisense phosphorodiamidate morpholino oligomers (PMO) to reduce myostatin expression in skeletal muscle and measured their effects on muscle mass and glucose uptake. C57/Bl6 mice received intramuscular or intravenous injections of anti-myostatin PMOs. Repeated intramuscular administration lead to a reduction in myostatin transcript levels (~ 20-40%), and an increase in muscle mass in chow and high-fat diet (HFD)-fed mice, but insulin-stimulated glucose uptake was reduced in PMO-treated muscles of HFD-fed mice. Five weekly intravenous administrations of 100 nmol PMO did not reduce myostatin expression, and therefore had no significant physiological effects. Unexpectedly, exon skipping levels were higher after intramuscular administration of PMO in HFD- than chow-fed mice. These results suggest that a modest PMO-induced reduction in myostatin transcript levels is sufficient to induce an increase in muscle mass, but that a greater degree of inhibition may be required to improve muscle glucose uptake.


Subject(s)
Insulin Resistance , Morpholinos/administration & dosage , Myostatin/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Exons , Glucose/metabolism , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL , Morpholinos/metabolism , Muscle, Skeletal/metabolism , Myostatin/antagonists & inhibitors , Myostatin/genetics
2.
J Physiol ; 599(3): 963-979, 2021 02.
Article in English | MEDLINE | ID: mdl-33258480

ABSTRACT

KEY POINTS: Reduced vitamin D receptor (VDR) expression prompts skeletal muscle atrophy. Atrophy occurs through catabolic processes, namely the induction of autophagy, while anabolism remains unchanged. In response to VDR-knockdown mitochondrial function and related gene-set expression is impaired. In vitro VDR knockdown induces myogenic dysregulation occurring through impaired differentiation. These results highlight the autonomous role the VDR has within skeletal muscle mass regulation. ABSTRACT: Vitamin D deficiency is estimated to affect ∼40% of the world's population and has been associated with impaired muscle maintenance. Vitamin D exerts its actions through the vitamin D receptor (VDR), the expression of which was recently confirmed in skeletal muscle, and its down-regulation is linked to reduced muscle mass and functional decline. To identify potential mechanisms underlying muscle atrophy, we studied the impact of VDR knockdown (KD) on mature skeletal muscle in vivo, and myogenic regulation in vitro in C2C12 cells. Male Wistar rats underwent in vivo electrotransfer (IVE) to knock down the VDR in hind-limb tibialis anterior (TA) muscle for 10 days. Comprehensive metabolic and physiological analysis was undertaken to define the influence loss of the VDR on muscle fibre composition, protein synthesis, anabolic and catabolic signalling, mitochondrial phenotype and gene expression. Finally, in vitro lentiviral transfection was used to induce sustained VDR-KD in C2C12 cells to analyse myogenic regulation. Muscle VDR-KD elicited atrophy through a reduction in total protein content, resulting in lower myofibre area. Activation of autophagic processes was observed, with no effect upon muscle protein synthesis or anabolic signalling. Furthermore, RNA-sequencing analysis identified systematic down-regulation of multiple mitochondrial respiration-related protein and genesets. Finally, in vitro VDR-knockdown impaired myogenesis (cell cycling, differentiation and myotube formation). Together, these data indicate a fundamental regulatory role of the VDR in the regulation of myogenesis and muscle mass, whereby it acts to maintain muscle mitochondrial function and limit autophagy.


Subject(s)
Receptors, Calcitriol , Vitamin D Deficiency , Animals , Male , Muscle Fibers, Skeletal , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Rats , Rats, Wistar , Receptors, Calcitriol/genetics , Vitamin D
3.
Mol Metab ; 42: 101059, 2020 12.
Article in English | MEDLINE | ID: mdl-32771696

ABSTRACT

OBJECTIVE: The Vitamin D receptor (VDR) has been positively associated with skeletal muscle mass, function and regeneration. Mechanistic studies have focused on the loss of the receptor, with in vivo whole-body knockout models demonstrating reduced myofibre size and function and impaired muscle development. To understand the mechanistic role upregulation of the VDR elicits in muscle mass/health, we studied the impact of VDR over-expression (OE) in vivo before exploring the importance of VDR expression upon muscle hypertrophy in humans. METHODS: Wistar rats underwent in vivo electrotransfer (IVE) to overexpress the VDR in the Tibialis anterior (TA) muscle for 10 days, before comprehensive physiological and metabolic profiling to characterise the influence of VDR-OE on muscle protein synthesis (MPS), anabolic signalling and satellite cell activity. Stable isotope tracer (D2O) techniques were used to assess sub-fraction protein synthesis, alongside RNA-Seq analysis. Finally, human participants underwent 20 wks of resistance exercise training, with body composition and transcriptomic analysis. RESULTS: Muscle VDR-OE yielded total protein and RNA accretion, manifesting in increased myofibre area, i.e., hypertrophy. The observed increases in MPS were associated with enhanced anabolic signalling, reflecting translational efficiency (e.g., mammalian target of rapamycin (mTOR-signalling), with no effects upon protein breakdown markers being observed. Additionally, RNA-Seq illustrated marked extracellular matrix (ECM) remodelling, while satellite cell content, markers of proliferation and associated cell-cycled related gene-sets were upregulated. Finally, induction of VDR mRNA correlated with muscle hypertrophy in humans following long-term resistance exercise type training. CONCLUSION: VDR-OE stimulates muscle hypertrophy ostensibly via heightened protein synthesis, translational efficiency, ribosomal expansion and upregulation of ECM remodelling-related gene-sets. Furthermore, VDR expression is a robust marker of the hypertrophic response to resistance exercise in humans. The VDR is a viable target of muscle maintenance through testable Vitamin D molecules, as active molecules and analogues.


Subject(s)
Hypertrophy/metabolism , Muscle, Skeletal/metabolism , Receptors, Calcitriol/metabolism , Adult , Animals , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Healthy Volunteers , Humans , Male , Middle Aged , Muscle Proteins/genetics , Myoblasts/metabolism , Myofibrils/metabolism , Physical Conditioning, Animal/methods , Rats , Rats, Wistar , Receptors, Calcitriol/genetics , Resistance Training/methods , Signal Transduction , Vitamin D/metabolism
4.
Am J Physiol Endocrinol Metab ; 319(1): E163-E174, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32459523

ABSTRACT

Myostatin inhibition is thought to improve whole body insulin sensitivity and mitigate the development of insulin resistance in models of obesity. However, although myostatin is known to be a major regulator of skeletal muscle mass, the direct effects of myostatin inhibition in muscle on glucose uptake and the mechanisms that may underlie this are still unclear. We investigated the effect of local myostatin inhibition by adeno-associated virus-mediated overexpression of the myostatin propeptide on insulin-stimulated skeletal muscle glucose disposal in chow-fed or high fat diet-fed mice and evaluated the molecular pathways that might mediate this. We found that myostatin inhibition improved glucose disposal in obese high fat diet-fed mice alongside the induction of muscle hypertrophy but did not have an impact in chow-fed mice. This improvement was not associated with greater glucose transporter or peroxisome proliferator-activated receptor-γ coactivator-1α expression or 5' AMP-activated protein kinase activation as previously suggested. Instead, transcriptomic analysis suggested that the improvement in glucose disposal was associated with significant enrichment in genes involved in fatty acid metabolism and translation of mitochondrial genes. Thus, myostatin inhibition improves muscle insulin-stimulated glucose disposal in obese high fat diet-fed mice independent of muscle hypertrophy, potentially involving previously unidentified pathways.


Subject(s)
Diet, High-Fat , Glucose/metabolism , Insulin Resistance/genetics , Muscle, Skeletal/metabolism , Myostatin/genetics , Protein Precursors/genetics , Animals , Dependovirus/genetics , Fatty Acids/metabolism , Gene Expression Profiling , Genes, Mitochondrial , Glucose Tolerance Test , Hypertrophy , Lipid Metabolism/genetics , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Myostatin/antagonists & inhibitors , Myostatin/metabolism , Obesity/metabolism , Protein Biosynthesis/genetics , Transfection
5.
Article in English | MEDLINE | ID: mdl-29209277

ABSTRACT

Type 2 diabetes mellitus (T2DM) is associated with skeletal complications, including an increased risk of fractures. Reduced blood supply and bone strength may contribute to this skeletal fragility. We hypothesized that long-term administration of Exenatide, a glucagon-like peptide-1 receptor agonist, would improve bone architecture and strength of T2DM mice by increasing blood flow to bone, thereby stimulating bone formation. In this study, we used a model of obesity and severe T2DM, the leptin receptor-deficient db/db mouse to assess alterations in bone quality and hindlimb blood flow and to examine the beneficial effects of 4 weeks administration of Exenatide. As expected, diabetic mice showed marked alterations in bone structure, remodeling and strength, and basal vascular tone compared with lean mice. Exenatide treatment improved trabecular bone mass and architecture by increasing bone formation rate, but only in diabetic mice. Although there was no effect on hindlimb perfusion at the end of this treatment, Exenatide administration acutely increased tibial blood flow. While Exenatide treatment did not restore the impaired bone strength, intrinsic properties of the matrix, such as collagen maturity, were improved. The effects of Exenatide on in vitro bone formation were further investigated in primary osteoblasts cultured under high-glucose conditions, showing that Exenatide reversed the impairment in bone formation induced by glucose. In conclusion, Exenatide improves trabecular bone mass by increasing bone formation and could protect against the development of skeletal complications associated with T2DM.

6.
Exp Physiol ; 101(6): 731-42, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27164183

ABSTRACT

What is the central question of this study? Although SGLT2 inhibitors represent a promising treatment for patients suffering from diabetic nephropathy, the influence of metabolic disruption on the expression and function of glucose transporters is largely unknown. What is the main finding and its importance? In vivo models of metabolic disruption (Goto-Kakizaki type II diabetic rat and junk-food diet) demonstrate increased expression of SGLT1, SGLT2 and GLUT2 in the proximal tubule brush border. In the type II diabetic model, this is accompanied by increased SGLT- and GLUT-mediated glucose uptake. A fasted model of metabolic disruption (high-fat diet) demonstrated increased GLUT2 expression only. The differential alterations of glucose transporters in response to varying metabolic stress offer insight into the therapeutic value of inhibitors. SGLT2 inhibitors are now in clinical use to reduce hyperglycaemia in type II diabetes. However, renal glucose reabsorption across the brush border membrane (BBM) is not completely understood in diabetes. Increased consumption of a Western diet is strongly linked to type II diabetes. This study aimed to investigate the adaptations that occur in renal glucose transporters in response to experimental models of diet-induced insulin resistance. The study used Goto-Kakizaki type II diabetic rats and normal rats rendered insulin resistant using junk-food or high-fat diets. Levels of protein kinase C-ßI (PKC-ßI), GLUT2, SGLT1 and SGLT2 were determined by Western blotting of purified renal BBM. GLUT- and SGLT-mediated d-[(3) H]glucose uptake by BBM vesicles was measured in the presence and absence of the SGLT inhibitor phlorizin. GLUT- and SGLT-mediated glucose transport was elevated in type II diabetic rats, accompanied by increased expression of GLUT2, its upstream regulator PKC-ßI and SGLT1 protein. Junk-food and high-fat diet feeding also caused higher membrane expression of GLUT2 and its upstream regulator PKC-ßI. However, the junk-food diet also increased SGLT1 and SGLT2 levels at the proximal tubule BBM. Glucose reabsorption across the proximal tubule BBM, via GLUT2, SGLT1 and SGLT2, is not solely dependent on glycaemic status, but is also influenced by diet-induced changes in glucose metabolism. We conclude that different metabolic disturbances result in complex adaptations in renal glucose transporter protein levels and function.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Kidney Tubules, Proximal/metabolism , Membranes/metabolism , Animals , Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Glucose/metabolism , Glucose Transporter Type 2/metabolism , Hyperglycemia/metabolism , Insulin Resistance/physiology , Kidney/metabolism , Male , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2/metabolism
7.
J Endocrinol ; 229(2): R67-81, 2016 05.
Article in English | MEDLINE | ID: mdl-26931135

ABSTRACT

Insulin resistance (IR) in skeletal muscle is a key defect mediating the link between obesity and type 2 diabetes, a disease that typically affects people in later life. Sarcopenia (age-related loss of muscle mass and quality) is a risk factor for a number of frailty-related conditions that occur in the elderly. In addition, a syndrome of 'sarcopenic obesity' (SO) is now increasingly recognised, which is common in older people and is applied to individuals that simultaneously show obesity, IR and sarcopenia. Such individuals are at an increased risk of adverse health events compared with those who are obese or sarcopenic alone. However, there are no licenced treatments for sarcopenia or SO, the syndrome is poorly defined clinically and the mechanisms that might explain a common aetiology are not yet well characterised. In this review, we detail the nature and extent of the clinical syndrome, highlight some of the key physiological processes that are dysregulated and discuss some candidate molecular pathways that could be implicated in both metabolic and anabolic defects in skeletal muscle, with an eye towards future therapeutic options. In particular, the potential roles of Akt/mammalian target of rapamycin signalling, AMP-activated protein kinase, myostatin, urocortins and vitamin D are discussed.


Subject(s)
Insulin Resistance/physiology , Sarcopenia/metabolism , Adipocytes/metabolism , Aged , Comorbidity , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Female , Glucose/metabolism , Humans , Lipid Metabolism , Male , Models, Biological , Muscle Proteins/metabolism , Obesity/epidemiology , Obesity/metabolism , Sarcopenia/epidemiology , Signal Transduction
9.
Diabetes ; 64(6): 1914-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25576050

ABSTRACT

Insulin and exercise stimulate glucose uptake into skeletal muscle via different pathways. Both stimuli converge on the translocation of the glucose transporter GLUT4 from intracellular vesicles to the cell surface. Two Rab guanosine triphosphatases-activating proteins (GAPs) have been implicated in this process: AS160 for insulin stimulation and its homolog, TBC1D1, are suggested to regulate exercise-mediated glucose uptake into muscle. TBC1D1 has also been implicated in obesity in humans and mice. We investigated the role of TBC1D1 in glucose metabolism by generating TBC1D1(-/-) mice and analyzing body weight, insulin action, and exercise. TBC1D1(-/-) mice showed normal glucose and insulin tolerance, with no difference in body weight compared with wild-type littermates. GLUT4 protein levels were reduced by ∼40% in white TBC1D1(-/-) muscle, and TBC1D1(-/-) mice showed impaired exercise endurance together with impaired exercise-mediated 2-deoxyglucose uptake into white but not red muscles. These findings indicate that the RabGAP TBC1D1 plays a key role in regulating GLUT4 protein levels and in exercise-mediated glucose uptake in nonoxidative muscle fibers.


Subject(s)
Muscle, Skeletal/metabolism , Nuclear Proteins/metabolism , Animals , Body Weight/genetics , Body Weight/physiology , Electrophoresis, Polyacrylamide Gel , Electroporation , GTPase-Activating Proteins , Glucose Transporter Type 4/metabolism , Glycogen/metabolism , Insulin/metabolism , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Physical Conditioning, Animal , Real-Time Polymerase Chain Reaction
10.
J Endocrinol ; 223(2): 143-54, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25122003

ABSTRACT

Insulin resistance (IR) in skeletal muscle is an important component of both type 2 diabetes and the syndrome of sarcopaenic obesity, for which there are no effective therapies. Urocortins (UCNs) are not only well established as neuropeptides but also have their roles in metabolism in peripheral tissues. We have shown recently that global overexpression of UCN3 resulted in muscular hypertrophy and resistance to the adverse metabolic effects of a high-fat diet. Herein, we aimed to establish whether short-term local UCN3 expression could enhance glucose disposal and insulin signalling in skeletal muscle. UCN3 was found to be expressed in right tibialis cranialis and extensor digitorum longus muscles of rats by in vivo electrotransfer and the effects studied vs the contralateral muscles after 1 week. No increase in muscle mass was detected, but test muscles showed 19% larger muscle fibre diameter (P=0.030), associated with increased IGF1 and IGF1 receptor mRNA and increased SER256 phosphorylation of forkhead transcription factor. Glucose clearance into the test muscles after an intraperitoneal glucose load was increased by 23% (P=0.018) per unit mass, associated with increased GLUT1 (34% increase; P=0.026) and GLUT4 (48% increase; P=0.0009) proteins, and significantly increased phosphorylation of insulin receptor substrate-1, AKT, AKT substrate of 160 kDa, glycogen synthase kinase-3ß, AMP-activated protein kinase and its substrate acetyl coA carboxylase. Thus, UCN3 expression enhances glucose disposal and signalling in muscle by an autocrine/paracrine mechanism that is separate from its pro-hypertrophic effects, implying that such a manipulation may have promised for the treatment of IR syndromes including sarcopaenic obesity.


Subject(s)
Adenylate Kinase/metabolism , Glucose/metabolism , Muscle, Skeletal/metabolism , Oncogene Protein v-akt/metabolism , Urocortins/physiology , Animals , Autocrine Communication/genetics , Male , Mice , Paracrine Communication/genetics , Rats , Rats, Transgenic , Rats, Wistar , Signal Transduction/genetics , Up-Regulation/genetics
11.
Mol Cell Endocrinol ; 393(1-2): 129-42, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24973767

ABSTRACT

Obesity and saturated fatty acid (SFA) treatment are both associated with skeletal muscle insulin resistance (IR) and increased macrophage infiltration. However, the relative effects of SFA and unsaturated fatty acid (UFA)-activated macrophages on muscle are unknown. Here, macrophages were treated with palmitic acid, palmitoleic acid or both and the effects of the conditioned medium (CM) on C2C12 myotubes investigated. CM from palmitic acid-treated J774s (palm-mac-CM) impaired insulin signalling and insulin-stimulated glycogen synthesis, reduced Inhibitor κBα and increased phosphorylation of p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase in myotubes. p38 MAPK inhibition or siRNA partially ameliorated these defects, as did addition of tumour necrosis factor-α blocking antibody to the CM. Macrophages incubated with both FAs generated CM that did not induce IR, while palmitoleic acid-mac-CM alone was insulin sensitising. Thus UFAs may improve muscle insulin sensitivity and counteract SFA-mediated IR through an effect on macrophage activation.


Subject(s)
Fatty Acids, Monounsaturated/pharmacology , Insulin Resistance , Macrophage Activation/drug effects , Muscle, Skeletal/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Myoblasts/cytology , Myoblasts/drug effects , Palmitic Acid/toxicity , Real-Time Polymerase Chain Reaction
12.
Am J Physiol Endocrinol Metab ; 304(8): E885-94, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23482445

ABSTRACT

Failure of white adipose tissue to appropriately store excess metabolic substrate seems to underpin obesity-associated type 2 diabetes. Encouraging "browning" of white adipose has been suggested as a therapeutic strategy to help dispose of excess stored lipid and ameliorate the resulting insulin resistance. Genetic variation at the DNA locus encoding the novel proteolipid neuronatin has been associated with obesity, and we recently observed that neuronatin expression is reduced in subcutaneous adipose tissue from obese humans. Thus, to explore the function of neuronatin further, we used RNAi to silence its expression in murine primary adipocyte cultures and examined the effects on adipocyte phenotype. We found that primary adipocytes express only the longer isoform of neuronatin. Loss of neuronatin led to increased mitochondrial biogenesis, indicated by greater intensity of MitoTracker Green staining. This was accompanied by increased expression of UCP1 and the key genes in mitochondrial oxidative phosphorylation, PGC-1α, Cox8b, and Cox4 in primary subcutaneous white adipocytes, indicative of a "browning" effect. In addition, phosphorylation of AMPK and ACC was increased, suggestive of increased fatty acid utilization. Similar, but less pronounced, effects of neuronatin silencing were also noted in primary brown adipocytes. In contrast, loss of neuronatin caused a reduction in both basal and insulin-stimulated glucose uptake and glycogen synthesis, likely mediated by a reduction in Glut1 protein upon silencing of neuronatin. In contrast, loss of neuronatin had no effect on insulin signaling. In conclusion, neuronatin appears to be a novel regulator of browning and metabolic substrate disposal in white adipocytes.


Subject(s)
Adipocytes, White/physiology , Adipose Tissue, Brown/physiology , Blood Glucose/metabolism , Glucose Transporter Type 1/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Obesity/genetics , Adipocytes, White/cytology , Adipogenesis/physiology , Adipose Tissue, Brown/cytology , Adult , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Glycogen/biosynthesis , Humans , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Middle Aged , Mitochondria/physiology , Nerve Tissue Proteins/metabolism , Obesity/metabolism , Obesity/physiopathology , Phenotype , Primary Cell Culture
13.
Am J Physiol Endocrinol Metab ; 303(6): E798-805, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22829583

ABSTRACT

Elevated mitochondrial reactive oxygen species have been suggested to play a causative role in some forms of muscle insulin resistance. However, the extent of their involvement in the development of diet-induced insulin resistance remains unclear. To investigate, manganese superoxide dismutase (MnSOD), a key mitochondrial-specific enzyme with antioxidant modality, was overexpressed, and the effect on in vivo muscle insulin resistance induced by a high-fat (HF) diet in rats was evaluated. Male Wistar rats were maintained on chow or HF diet. After 3 wk, in vivo electroporation (IVE) of MnSOD expression and empty vectors was undertaken in right and left tibialis cranialis (TC) muscles, respectively. After one more week, insulin action was evaluated using hyperinsulinemic euglycemic clamp, and tissues were subsequently analyzed for antioxidant enzyme capacity and markers of oxidative stress. MnSOD mRNA was overexpressed 4.5-fold, and protein levels were increased by 70%, with protein detected primarily in the mitochondrial fraction of muscle fibers. This was associated with elevated MnSOD and glutathione peroxidase activity, indicating that the overexpressed MnSOD was functionally active. The HF diet significantly reduced whole body and TC muscle insulin action, whereas overexpression of MnSOD in HF diet animals ameliorated this reduction in TC muscle glucose uptake by 50% (P < 0.05). Decreased protein carbonylation was seen in MnSOD overexpressing TC muscle in HF-treated animals (20% vs. contralateral control leg, P < 0.05), suggesting that this effect was mediated through an altered redox state. Thus interventions causing elevation of mitochondrial antioxidant activity may offer protection against diet-induced insulin resistance in skeletal muscle.


Subject(s)
Diet, High-Fat/adverse effects , Insulin Resistance , Muscle, Skeletal/enzymology , Oxidative Stress , Superoxide Dismutase/metabolism , Up-Regulation , Animals , Electroporation , Gene Transfer Techniques , Glutathione Peroxidase/metabolism , Humans , Lower Extremity , Male , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Protein Carbonylation , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recombinant Fusion Proteins/metabolism , Superoxide Dismutase/genetics
14.
Biochem Biophys Res Commun ; 388(4): 654-9, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19682428

ABSTRACT

The nuclear hormone receptor, Rev-erb beta operates as a transcriptional silencer. We previously demonstrated that exogenous expression of Rev-erb betaDeltaE in skeletal muscle cells increased Srebp-1c mRNA expression. We validated these in vitro observations by injection of an expression vector driving Rev-erb betaDeltaE expression into mouse tibialis muscle that resulted in increased Srebp-1c mRNA expression. Paradoxically, Rev-erb beta siRNA expression in skeletal muscle cells repressed Srebp-1c expression, and indicated that Rev-erb beta expression was necessary for Srebp-1c expression. ChIP analysis demonstrated that Rev-erb beta was recruited to the Srebp-1c promoter. Moreover, Rev-erb beta trans-activated the Srebp-1c promoter, in contrast, Rev-erb beta efficiently repressed the Rev-erb alpha promoter, a previously characterized target gene. Finally, treatment with the Rev-erb agonist (hemin) (i) increased the trans-activation of the Srebp-1c promoter by Rev-erb beta; and (ii) increased Rev-erb beta and Srebp-1c mRNA expression. These data suggest that Rev-erb beta has the potential to activate gene expression, and is a positive regulator of Srebp-1c, a regulator of lipogenesis.


Subject(s)
Muscle, Skeletal/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Transcriptional Activation , Animals , Base Sequence , Cell Line , Electroporation , Hemin/pharmacology , Hindlimb , Lipogenesis/genetics , Mice , Molecular Sequence Data , Muscle, Skeletal/cytology , Promoter Regions, Genetic/drug effects , RNA, Messenger/biosynthesis , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/agonists , Repressor Proteins/genetics
15.
Am J Physiol Endocrinol Metab ; 294(2): E316-25, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18029440

ABSTRACT

Insulin resistance of skeletal muscle is a major defect in obesity and type 2 diabetes. Insulin resistance has been associated with a chronic subclinical inflammatory state in epidemiological studies and specifically with activation of the inhibitor kappaB kinase (IkappaBK)-nuclear factor-kappaB (NF-kappaB) pathway. However, it is unclear whether this pathway plays a role in mediating insulin resistance in muscle in vivo. We separately overexpressed the p65 subunit of NF-kappaB and IkappaBKbeta in single muscles of rats using in vivo electrotransfer and compared the effects after 1 wk vs. paired contralateral control muscles. A 64% increase in p65 protein (P < 0.001) was sufficient to cause muscle fiber atrophy but had no effect on glucose disposal or glycogen storage in muscle under hyperinsulinemic-euglycemic clamp conditions. Similarly, a 650% increase in IkappaBKbeta expression (P < 0.001) caused a significant reduction in IkappaB protein but also had no effect on clamp glucose disposal after lipid infusion. In fact, IkappaBKbeta overexpression in particular caused increases in activating tyrosine phosphorylation of insulin receptor substrate-1 (24%; P = 0.02) and serine phosphorylation of Akt (23%; P < 0.001), implying a moderate increase in flux through the insulin signaling cascade. Interestingly, p65 overexpression resulted in a negative feedback reduction of 36% in Toll-like receptor (TLR)-2 (P = 0.03) but not TLR-4 mRNA. In conclusion, activation of the IkappaBKbeta-NF-kappaB pathway in muscle does not seem to be an important local mediator of insulin resistance.


Subject(s)
I-kappa B Kinase/physiology , Insulin Resistance/physiology , NF-kappa B/physiology , Signal Transduction/physiology , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Feedback, Physiological , Gene Transfer Techniques , Genetic Vectors , Glucose/metabolism , Glucose Clamp Technique , Immunohistochemistry , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/metabolism
16.
Mol Endocrinol ; 21(1): 215-28, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17021050

ABSTRACT

The phosphoinositide 3-kinase/Akt pathway is thought to be essential for normal insulin action and glucose metabolism in skeletal muscle and has been shown to be dysregulated in insulin resistance. However, the specific roles of and signaling pathways triggered by Akt isoforms have not been fully assessed in muscle in vivo. We overexpressed constitutively active (ca-) Akt-1 or Akt-2 constructs in muscle using in vivo electrotransfer and, after 1 wk, assessed the roles of each isoform on glucose metabolism and fiber growth. We achieved greater than 2.5-fold increases in total Ser473 phosphorylation in muscles expressing ca-Akt-1 and ca-Akt-2, respectively. Both isoforms caused hypertrophy of muscle fibers, consistent with increases in p70S6kinase phosphorylation, and a 60% increase in glycogen accumulation, although only Akt-1 increased glycogen synthase kinase-3beta phosphorylation. Akt-2, but not Akt-1, increased basal glucose uptake (by 33%, P = 0.004) and incorporation into glycogen and lipids, suggesting a specific effect on glucose transport. Consistent with this, short hairpin RNA-mediated silencing of Akt-2 caused reductions in glycogen storage and glucose uptake. Consistent with Akt-mediated insulin receptor substrate 1 (IRS-1) degradation, we observed approximately 30% reductions in IRS-1 protein in muscle overexpressing ca-Akt-1 or ca-Akt-2. Despite this, we observed no decrease in insulin-stimulated glucose uptake. Furthermore, a 68% reduction in IRS-1 levels induced using short hairpin RNAs targeting IRS-1 also did not affect glucose disposal after a glucose load. These data indicate distinct roles for Akt-1 and Akt-2 in muscle glucose metabolism and that moderate reductions in IRS-1 expression do not result in the development of insulin resistance in skeletal muscle in vivo.


Subject(s)
Gene Expression Regulation, Enzymologic , Insulin/metabolism , Muscle, Skeletal/metabolism , Phosphoproteins/biosynthesis , Proto-Oncogene Proteins c-akt/physiology , Animals , Deoxyglucose/chemistry , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin Receptor Substrate Proteins , Mice , Models, Biological , Phosphorylation , Protein Isoforms , Proto-Oncogene Proteins c-akt/chemistry , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
17.
Am J Physiol Endocrinol Metab ; 292(4): E1231-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17179390

ABSTRACT

A key regulatory point in the control of fatty acid (FA) oxidation is thought to be transport of FAs across the mitochondrial membrane by carnitine palmitoyltransferase I (CPT I). To investigate the role of CPT I in FA metabolism, we used in vivo electrotransfer (IVE) to locally overexpress CPT I in muscle of rodents. A vector expressing the human muscle isoform of CPT I was electrotransferred into the right lateral muscles of the distal hindlimb [tibialis cranialis (TC) and extensor digitorum longus (EDL)] of rats, and a control vector expressing GFP was electrotransferred into the left muscles. Initial studies showed that CPT I protein expression peaked 7 days after IVE (+104%, P<0.01). This was associated with an increase in maximal CPT I activity (+30%, P < 0.001) and a similar increase in palmitoyl-CoA oxidation (+24%; P<0.001) in isolated mitochondria from the TC. Importantly, oxidation of the medium-chain FA octanoyl-CoA and CPT I sensitivity to inhibition by malonyl-CoA were not altered by CPT I overexpression. FA oxidation in isolated EDL muscle strips was increased with CPT I overexpression (+28%, P<0.01), whereas FA incorporation into the muscle triacylglycerol (TAG) pool was reduced (-17%, P<0.01). As a result, intramyocellular TAG content was decreased with CPT I overexpression in both the TC (-25%, P<0.05) and the EDL (-45%, P<0.05). These studies demonstrate that acute overexpression of CPT I in muscle leads to a repartitioning of FAs away from esterification and toward oxidation and highlight the importance of CPT I in regulating muscle FA metabolism.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Fatty Acids/metabolism , Muscle, Skeletal/metabolism , Triglycerides/metabolism , Animals , Biomarkers/metabolism , Carnitine O-Palmitoyltransferase/genetics , Electroporation , Esterification , Hindlimb , Humans , In Vitro Techniques , Lipid Metabolism , Male , Mitochondria, Muscle/enzymology , Muscle, Skeletal/enzymology , Oxidation-Reduction , Palmitates/metabolism , Palmitoyl Coenzyme A/metabolism , Rats , Rats, Wistar , Transfection/methods
18.
Diabetes ; 54(9): 2702-11, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123360

ABSTRACT

Analysis of conventional germ-line or tissue-specific gene manipulation in vivo is potentially confounded by developmental adaptation of animal physiology. We aimed to adapt the technique of in vivo electrotransfer (IVE) to alter local gene expression in skeletal muscle of rodents as a means of investigating the role of specific proteins in glucose metabolism in vivo. We utilized a square-wave electroporator to induce intracellular electrotransfer of DNA constructs injected into rat or mouse muscles and investigated the downstream effects. In initial studies, expression of green fluorescent protein reporter was induced in 53 +/- 10% of muscle fibers peaking at 7 days, and importantly, the electrotransfer procedure itself did not impact upon the expression of stress proteins or our ability to detect a reduction in 2-deoxyglucose tracer uptake by electroporated muscle of high-fat-fed rats during hyperinsulinemic-euglycemic clamp. To demonstrate functional effects of electrotransfer of constructs targeting glucose transporters, we administered vectors encoding GLUT-1 cDNA and GLUT-4 short hairpin RNAs (shRNAs) to rodent muscles. IVE of the GLUT-1 gene resulted in a 57% increase in GLUT-1 protein, accompanied by a proportionate increase in basal 2-deoxyglucose tracer uptake into muscles of starved rats. IVE of vectors expressing two shRNAs for GLUT-4 demonstrated to reduce specific protein expression and 2-deoxyglucose tracer uptake in 3T3-L1 adipocytes into mouse muscle caused a 51% reduction in GLUT-4 protein, associated with attenuated clearance of tracer to muscle after a glucose load. These results confirm that glucose transporter expression is largely rate limiting for glucose uptake in vivo and highlight the utility of IVE for the acute manipulation of muscle gene expression in the study of the role of specific proteins in glucose metabolism.


Subject(s)
Glucose/metabolism , Muscle Proteins/metabolism , Animals , Biological Transport, Active , Cell Line , Electroporation , Gene Expression Regulation , Male , Mice , Rats , Rats, Wistar
19.
J Biol Chem ; 280(13): 12573-84, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15640143

ABSTRACT

Skeletal muscle is a major mass peripheral tissue that accounts for approximately 40% of total body weight and 50% of energy expenditure and is a primary site of glucose disposal and fatty acid oxidation. Consequently, muscle has a significant role in insulin sensitivity, obesity, and the blood-lipid profile. Excessive caloric intake is sensed by the brain and induces beta-adrenergic receptor (beta-AR)-mediated adaptive thermogenesis. Beta-AR null mice develop severe obesity on a high fat diet. However, the target gene(s), target tissues(s), and molecular mechanism involved remain obscure. We observed that 30-60 min of beta-AR agonist (isoprenaline) treatment of C2C12 skeletal muscle cells strikingly activated (>100-fold) the expression of the mRNA encoding the nuclear hormone receptor, Nur77. In contrast, the expression of other nuclear receptors that regulate lipid and carbohydrate metabolism was not induced. Stable transfection of Nur77-specific small interfering RNAs (siNur77) into skeletal muscle cells repressed endogenous Nur77 mRNA expression. Moreover, we observed attenuation of gene and protein expression associated with the regulation of energy expenditure and lipid homeostasis, for example AMP-activated protein kinase gamma3, UCP3, CD36, adiponectin receptor 2, GLUT4, and caveolin-3. Attenuation of Nur77 expression resulted in decreased lipolysis. Finally, in concordance with the cell culture model, injection and electrotransfer of siNur77 into mouse tibialis cranialis muscle resulted in the repression of UCP3 mRNA expression. This study demonstrates regulatory cross-talk between the nuclear hormone receptor and beta-AR signaling pathways. Moreover, it suggests Nur77 modulates the expression of genes that are key regulators of skeletal muscle lipid and energy homeostasis. In conclusion, we speculate that Nur77 agonists would stimulate lipolysis and increase energy expenditure in skeletal muscle and suggest selective activators of Nur77 may have therapeutic utility in the treatment of obesity.


Subject(s)
DNA-Binding Proteins/physiology , Lipid Metabolism , Muscle, Skeletal/cytology , Receptors, Adrenergic, beta/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Steroid/physiology , Transcription Factors/physiology , Animals , Blotting, Western , Carbohydrate Metabolism , Cell Line , Cell Nucleus/metabolism , DNA Primers/chemistry , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , Electroporation , Gene Expression Regulation , Glucose/metabolism , Hot Temperature , Mice , Models, Biological , Muscle, Skeletal/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1 , Plasmids/metabolism , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription Factors/metabolism , Transfection
20.
Diabetes ; 53(12): 3258-66, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15561958

ABSTRACT

Metformin reduces the incidence of progression to type 2 diabetes in humans with obesity or impaired glucose tolerance. We used an animal model to investigate whether metformin could prevent acute lipid-induced insulin resistance and the mechanisms involved. Metformin or vehicle was administered to rats daily for 1 week. Rats were studied basally, after 3.75 h of intralipid-heparin or glycerol infusion, or after 5 h of infusion with a hyperinsulinemic-euglycemic clamp between 3 and 5 h. Metformin had no effect on plasma triacylglycerol or nonesterified fatty acid concentrations and did not alter glucose turnover or gluconeogenic enzyme mRNA after lipid infusion. However, metformin normalized hepatic glucose output and increased liver glycogen during lipid infusion and clamp. Basal liver (but not muscle or fat) AMP-activated protein kinase activity was increased by metformin (by 310%; P < 0.01), associated with increased phosphorylation of acetyl CoA carboxylase. Postclamp liver but not muscle phosphorylated/total Akt protein was increased, whereas basal c-Jun NH2-terminal kinase-1 and -2 protein expression were reduced (by 39 and 53%, respectively; P < 0.05). Metformin also increased hepatic basal IkappaBalpha levels (by 260%; P < 0.001) but had no effect on tyrosine phosphorylation or expression of insulin receptor substrate-1 (IRS-1). In summary, metformin opposes the development of acute lipid-induced insulin resistance in the liver through alterations in multiple signaling pathways.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Lipids/pharmacology , Liver/physiology , Metformin/pharmacology , Signal Transduction/drug effects , Animals , Gene Expression Regulation/drug effects , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Glucose/metabolism , Glucose Clamp Technique , Hyperinsulinism , Liver/drug effects , Male , Metabolic Syndrome/prevention & control , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...