Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
2.
Int J Mol Sci ; 20(7)2019 Mar 27.
Article in English | MEDLINE | ID: mdl-30934678

ABSTRACT

Adiponectin regulates metabolism through blood glucose control and fatty acid oxidation, partly mediated by downstream effects of adiponectin signaling in skeletal muscle. More recently, skeletal muscle has been identified as a source of adiponectin expression, fueling interest in the role of adiponectin as both a circulating adipokine and a locally expressed paracrine/autocrine factor. In addition to being metabolically responsive, skeletal muscle functional capacity, calcium handling, growth and maintenance, regenerative capacity, and susceptibility to chronic inflammation are all strongly influenced by adiponectin stimulation. Furthermore, physical exercise has clear links to adiponectin expression and circulating concentrations in healthy and diseased populations. Greater physical activity is generally related to higher adiponectin expression while lower adiponectin levels are found in inactive obese, pre-diabetic, and diabetic populations. Exercise training typically restores plasma adiponectin and is associated with improved insulin sensitivity. Thus, the role of adiponectin signaling in skeletal muscle has expanded beyond that of a metabolic regulator to include several aspects of skeletal muscle function and maintenance critical to muscle health, many of which are responsive to, and mediated by, physical exercise.


Subject(s)
Adiponectin/metabolism , Muscle, Skeletal/metabolism , Autophagy , Exercise/physiology , Humans , Protein Processing, Post-Translational , Regeneration/physiology
3.
Diabetologia ; 61(6): 1411-1423, 2018 06.
Article in English | MEDLINE | ID: mdl-29666899

ABSTRACT

AIMS/HYPOTHESIS: A comprehensive assessment of skeletal muscle ultrastructure and mitochondrial bioenergetics has not been undertaken in individuals with type 1 diabetes. This study aimed to systematically assess skeletal muscle mitochondrial phenotype in young adults with type 1 diabetes. METHODS: Physically active, young adults (men and women) with type 1 diabetes (HbA1c 63.0 ± 16.0 mmol/mol [7.9% ± 1.5%]) and without type 1 diabetes (control), matched for sex, age, BMI and level of physical activity, were recruited (n = 12/group) to undergo vastus lateralis muscle microbiopsies. Mitochondrial respiration (high-resolution respirometry), site-specific mitochondrial H2O2 emission and Ca2+ retention capacity (CRC) (spectrofluorometry) were assessed using permeabilised myofibre bundles. Electron microscopy and tomography were used to quantify mitochondrial content and investigate muscle ultrastructure. Skeletal muscle microvasculature was assessed by immunofluorescence. RESULTS: Mitochondrial oxidative capacity was significantly lower in participants with type 1 diabetes vs the control group, specifically at Complex II of the electron transport chain, without differences in mitochondrial content between groups. Muscles of those with type 1 diabetes also exhibited increased mitochondrial H2O2 emission at Complex III and decreased CRC relative to control individuals. Electron tomography revealed an increase in the size and number of autophagic remnants in the muscles of participants with type 1 diabetes. Despite this, levels of the autophagic regulatory protein, phosphorylated AMP-activated protein kinase (p-AMPKαThr172), and its downstream targets, phosphorylated Unc-51 like autophagy activating kinase 1 (p-ULK1Ser555) and p62, was similar between groups. In addition, no differences in muscle capillary density or platelet aggregation were observed between the groups. CONCLUSIONS/INTERPRETATION: Alterations in mitochondrial ultrastructure and bioenergetics are evident within the skeletal muscle of active young adults with type 1 diabetes. It is yet to be elucidated whether more rigorous exercise may help to prevent skeletal muscle metabolic deficiencies in both active and inactive individuals with type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Adult , Body Mass Index , Calcium/chemistry , Diabetes Mellitus, Type 1/pathology , Energy Metabolism , Exercise/physiology , Female , Humans , Hydrogen Peroxide/metabolism , Insulin/metabolism , Male , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Mitochondria/ultrastructure , Muscle, Skeletal/pathology , Oxygen Consumption , Young Adult
4.
FASEB J ; 28(5): 2098-107, 2014 May.
Article in English | MEDLINE | ID: mdl-24522207

ABSTRACT

AMP-activated protein kinase (AMPK) is a master regulator of metabolism. While muscle-specific AMPK ß1ß2 double-knockout (ß1ß2M-KO) mice display alterations in metabolic and mitochondrial capacity, their severe exercise intolerance suggested a secondary contributor to the observed phenotype. We find that tibialis anterior (TA), but not soleus, muscles of sedentary ß1ß2M-KO mice display a significant myopathy (decreased myofiber areas, increased split and necrotic myofibers, and increased centrally nucleated myofibers. A mitochondrial- and fiber-type-specific etiology to the myopathy was ruled out. However, ß1ß2M-KO TA muscles displayed significant (P<0.05) increases in platelet aggregation and apoptosis within myofibers and surrounding interstitium (P<0.05). These changes correlated with a 45% decrease in capillary density (P<0.05). We hypothesized that the ß1ß2M-KO myopathy in resting muscle resulted from impaired AMPK-nNOSµ signaling, causing increased platelet aggregation, impaired vasodilation, and, ultimately, ischemic injury. Consistent with this hypothesis, AMPK-specific phosphorylation (Ser1446) of nNOSµ was decreased in ß1ß2M-KO compared to wild-type (WT) mice. The AMPK-nNOSµ relationship was further demonstrated by administration of 5-aminoimidazole-4-carboxamide 1-ß-D-ribofuranoside (AICAR) to ß1ß2-MKO muscles and C2C12 myotubes. AICAR significantly increased nNOSµ phosphorylation and nitric oxide production (P<0.05) within minutes of administration in WT muscles and C2C12 myotubes but not in ß1ß2M-KO muscles. These findings highlight the importance of the AMPK-nNOSµ pathway in resting skeletal muscle.


Subject(s)
AMP-Activated Protein Kinases/genetics , Capillaries/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Nitric Oxide/metabolism , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Cell Line , Electron Transport Complex IV/metabolism , Female , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Muscle, Skeletal/blood supply , Necrosis/metabolism , Phosphorylation , Platelet Aggregation , Ribonucleotides/metabolism , Succinate Dehydrogenase/metabolism
5.
Pediatr Diabetes ; 16(1): 48-57, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24444038

ABSTRACT

BACKGROUND: Patients with type 1 diabetes mellitus (T1DM) may experience poor muscle health as a result of chronic hyperglycemia. Despite this, muscle function in children with T1DM with good or poor glycemic control has yet to be examined in detail. OBJECTIVE: To assess differences in muscle-related fitness variables in children with T1DM with good glycemic control (T1DM-G), as well as those with poor glycemic control (T1DM-P), and non-diabetic, healthy controls. SUBJECTS: Eight children with T1DM-G [glycosylated hemoglobin (HbA1c) ≤ 7.5% for 9 months], eight children with T1DM-P (HbA1c ≥ 9.0% for 9 months), and eight healthy controls completed one exercise session. METHODS: Anaerobic and aerobic muscle functions were assessed with a maximal isometric grip strength test, a Wingate test, and an incremental continuous cycling test until exhaustion. Blood samples were collected at rest to determine HbA1c at the time of testing. Physical activity was monitored over 7 d using accelerometry. RESULTS: Children with T1DM-P displayed lower peak oxygen consumption (VO2peak ) values (mL/kg/min) compared to healthy controls (T1DM-P: 33.2 ± 5.6, controls: 43.5 ± 6.3, p < 0.01), while T1DM-G (43.5 ± 6.3) had values similar to controls and T1DM-P. There was a negative relationship between VO2peak and HbA1c% (r = -0.54, p < 0.01). All groups were similar in all other fitness variables. There were no group differences in physical activity variables. CONCLUSION: Children with T1DM-G did not display signs of impaired muscle function, while children with T1DM-P have signs of altered aerobic muscle capacity.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/therapy , Motor Activity/physiology , Physical Fitness/physiology , Adolescent , Child , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Exercise/physiology , Female , Hand Strength , Heart Rate , Humans , Male , Muscle Strength , Oxygen Consumption , Pulmonary Gas Exchange/physiology
6.
J Appl Physiol (1985) ; 106(5): 1650-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19246652

ABSTRACT

Mechanistic studies examining the effects of Type 1 diabetes mellitus (T1DM) on skeletal muscle have largely relied on streptozotocin-induced diabetic (STZ) rodents. Unfortunately, characterization of diabetic myopathy in this model is confounded by the effects of streptozotocin on skeletal muscle independent of the diabetic phenotype. Here we define adolescent diabetic myopathy in a novel, genetic model of T1DM, Ins2(Akita+/-) mice, and contrast these findings with STZ mice. Eight weeks of diabetes resulted in significantly reduced gastrocnemius-plantaris-soleus mass (control: 0.16 +/- 0.005 g; Ins2(Akita+/-): 0.12 +/- 0.003 g; STZ: 0.12 +/- 0.01g) and IIB/D fiber area in Ins2(Akita+/-) (1,294 +/- 94 microm(2)) and STZ (1,768 +/- 163 microm(2)) compared with control (2,241 +/- 144 microm(2)). Conversely, STZ type I fibers (1,535 +/- 165 microm(2)) were significantly larger than Ins2(Akita+/-) (915 +/- 76 microm(2)) but not control (1,152 +/- 86 microm(2)). Intramyocellular lipid increased in STZ (122.9 +/- 3.6% of control) but not Ins2(Akita+/-) likely resultant from depressed citrate synthase (control: 6.2 +/- 1.2 micromol.s(-1).mg(-1); Ins2(Akita+/-): 5.2 +/- 0.8 micromol.s(-1).mg(-1); STZ: 2.8 +/- 0.5 micromol.s(-1).mg(-1)) and 3-beta-hydroxyacyl coenzyme-A dehydrogenase (control: 4.2 +/- 0.6 nmol.s(-1).mg(-1); Ins2(Akita+/-): 5.0 +/- 0.6 nmol.s(-1).mg(-1); STZ: 2.7 +/- 0.6 nmol.s(-1).mg(-1)) enzyme activity in STZ muscle. In situ muscle stimulation revealed lower absolute peak tetanic force in Ins2(Akita+/-) (70.2 +/- 8.2% of control) while STZ exhibited an insignificant decrease (87.6 +/- 7.9% of control). Corrected for muscle mass, no force loss was observed in Ins2(Akita+/-), while STZ was significantly elevated vs. control and Ins2(Akita+/-). These results demonstrate that atrophy and specific fiber-type loss in Ins2(Akita+/-) muscle did not affect contractile properties (relative to muscle mass). Furthermore, we demonstrate distinctive contractile, metabolic, and phenotypic properties in STZ vs. Ins2(Akita+/-) diabetic muscle despite similarity in hyperglycemia/hypoinsulinemia, raising concerns of our current state of knowledge regarding the effects of T1DM on skeletal muscle.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Muscle, Skeletal/pathology , Myositis/pathology , Animals , Diabetes Complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Electric Stimulation , Insulin/genetics , Insulin/metabolism , Lipids/biosynthesis , Male , Mice , Muscle Contraction/physiology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/drug effects , Myositis/chemically induced , Myositis/genetics
8.
Diabetes ; 65(10): 3053-61, 2016 10.
Article in English | MEDLINE | ID: mdl-27335233

ABSTRACT

Type 1 diabetes (T1D) negatively influences skeletal muscle health; however, its effect on muscle satellite cells (SCs) remains largely unknown. SCs from samples from rodents (Akita) and human subjects with T1D were examined to discern differences in SC density and functionality compared with samples from their respective control subjects. Examination of the Notch pathway was undertaken to investigate its role in changes to SC functionality. Compared with controls, Akita mice demonstrated increased muscle damage after eccentric exercise along with a decline in SC density and myogenic capacity. Quantification of components of the Notch signaling pathway revealed a persistent activation of Notch signaling in Akita SCs, which could be reversed with the Notch inhibitor DAPT. Similar to Akita samples, skeletal muscle from human subjects with T1D displayed a significant reduction in SC content, and the Notch ligand, DLL1, was significantly increased compared with control subjects, supporting the dysregulated Notch pathway observed in Akita muscles. These data indicate that persistent activation in Notch signaling impairs SC functionality in the T1D muscle, resulting in a decline in SC content. Given the vital role played by the SC in muscle growth and maintenance, these findings suggest that impairments in SC capacities play a primary role in the skeletal muscle myopathy that characterizes T1D.


Subject(s)
Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology , Adolescent , Adult , Animals , Cell Count , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Physical Conditioning, Animal/physiology , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction/physiology , Young Adult
9.
Cell Stem Cell ; 19(4): 433-448, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27376984

ABSTRACT

Adult mammals have lost multi-tissue regenerative capacity, except for the distal digit, which is able to regenerate via mechanisms that remain largely unknown. Here, we show that, after adult mouse distal digit removal, nerve-associated Schwann cell precursors (SCPs) dedifferentiate and secrete growth factors that promote expansion of the blastema and digit regeneration. When SCPs were dysregulated or ablated, mesenchymal precursor proliferation in the blastema was decreased and nail and bone regeneration were impaired. Transplantation of exogenous SCPs rescued these regeneration defects. We found that SCPs secrete factors that promote self-renewal of mesenchymal precursors, and we used transcriptomic and proteomic analysis to define candidate factors. Two of these, oncostatin M (OSM) and platelet-derived growth factor AA (PDGF-AA), are made by SCPs in the regenerating digit and rescued the deficits in regeneration caused by loss of SCPs. As all peripheral tissues contain nerves, these results could have broad implications for mammalian tissue repair and regeneration.


Subject(s)
Cell Dedifferentiation , Extremities/physiology , Mammals/physiology , Neural Stem Cells/cytology , Paracrine Communication , Regeneration , Schwann Cells/cytology , Aging/physiology , Animals , Cell Dedifferentiation/drug effects , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Denervation , Extremities/innervation , Gene Deletion , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/cytology , Mice , Mice, Knockout , Neural Stem Cells/transplantation , Oncostatin M/pharmacology , Paracrine Communication/drug effects , Platelet-Derived Growth Factor/pharmacology , Rats , Regeneration/drug effects , SOXB1 Transcription Factors/metabolism , Schwann Cells/transplantation , Skin/pathology , Wound Healing/drug effects
10.
Dev Cell ; 32(1): 31-42, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25556659

ABSTRACT

Ankrd11 is a potential chromatin regulator implicated in neural development and autism spectrum disorder (ASD) with no known function in the brain. Here, we show that knockdown of Ankrd11 in developing murine or human cortical neural precursors caused decreased proliferation, reduced neurogenesis, and aberrant neuronal positioning. Similar cellular phenotypes and aberrant ASD-like behaviors were observed in Yoda mice carrying a point mutation in the Ankrd11 HDAC-binding domain. Consistent with a role for Ankrd11 in histone acetylation, Ankrd11 was associated with chromatin and colocalized with HDAC3, and expression and histone acetylation of Ankrd11 target genes were altered in Yoda neural precursors. Moreover, the Ankrd11 knockdown-mediated decrease in precursor proliferation was rescued by inhibiting histone acetyltransferase activity or expressing HDAC3. Thus, Ankrd11 is a crucial chromatin regulator that controls histone acetylation and gene expression during neural development, thereby providing a likely explanation for its association with cognitive dysfunction and ASD.


Subject(s)
Autistic Disorder/pathology , Cell Proliferation , Chromatin/genetics , DNA-Binding Proteins/physiology , Histone Deacetylases/metabolism , Neurogenesis/genetics , Acetylation , Animals , Autistic Disorder/genetics , Autistic Disorder/metabolism , Behavior, Animal , Biomarkers/metabolism , Blotting, Western , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation , Female , Gene Expression Profiling , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Histones/metabolism , Immunoprecipitation , Mice , Oligonucleotide Array Sequence Analysis , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction
11.
Stem Cell Reports ; 3(1): 85-100, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25068124

ABSTRACT

Recent reports of directed reprogramming have raised questions about the stability of cell lineages. Here, we have addressed this issue, focusing upon skin-derived precursors (SKPs), a dermally derived precursor cell. We show by lineage tracing that murine SKPs from dorsal skin originate from mesenchymal and not neural crest-derived cells. These mesenchymally derived SKPs can, without genetic manipulation, generate functional Schwann cells, a neural crest cell type, and are highly similar at the transcriptional level to Schwann cells isolated from the peripheral nerve. This is not a mouse-specific phenomenon, since human SKPs that are highly similar at the transcriptome level can be made from neural crest-derived facial and mesodermally derived foreskin dermis and the foreskin SKPs can make myelinating Schwann cells. Thus, nonneural crest-derived mesenchymal precursors can differentiate into bona fide peripheral glia in the absence of genetic manipulation, suggesting that developmentally defined lineage boundaries are more flexible than widely thought.


Subject(s)
Multipotent Stem Cells/cytology , Schwann Cells/cytology , Skin/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Humans , Mice, SCID , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
12.
PLoS One ; 8(8): e70971, 2013.
Article in English | MEDLINE | ID: mdl-23951058

ABSTRACT

BACKGROUND: Systemic elevations in PAI-1 suppress the fibrinolytic pathway leading to poor collagen remodelling and delayed regeneration of tibialis anterior (TA) muscles in type-1 diabetic Akita mice. However, how impaired collagen remodelling was specifically attenuating regeneration in Akita mice remained unknown. Furthermore, given intrinsic differences between muscle groups, it was unclear if the reparative responses between muscle groups were different. PRINCIPAL FINDINGS: Here we reveal that diabetic Akita muscles display differential regenerative responses with the TA and gastrocnemius muscles exhibiting reduced regenerating myofiber area compared to wild-type mice, while soleus muscles displayed no difference between animal groups following injury. Collagen levels in TA and gastrocnemius, but not soleus, were significantly increased post-injury versus controls. At 5 days post-injury, when degenerating/necrotic regions were present in both animal groups, Akita TA and gastrocnemius muscles displayed reduced macrophage and satellite cell infiltration and poor myofiber formation. By 10 days post-injury, necrotic regions were absent in wild-type TA but persisted in Akita TA. In contrast, Akita soleus exhibited no impairment in any of these measures compared to wild-type soleus. In an effort to define how impaired collagen turnover was attenuating regeneration in Akita TA, a PAI-1 inhibitor (PAI-039) was orally administered to Akita mice following cardiotoxin injury. PAI-039 administration promoted macrophage and satellite cell infiltration into necrotic areas of the TA and gastrocnemius. Importantly, soleus muscles exhibit the highest inducible expression of MMP-9 following injury, providing a mechanism for normative collagen degradation and injury recovery in this muscle despite systemically elevated PAI-1. CONCLUSIONS: Our findings suggest the mechanism underlying how impaired collagen remodelling in type-1 diabetes results in delayed regeneration is an impairment in macrophage infiltration and satellite cell recruitment to degenerating areas; a phenomena that occurs differentially between muscle groups.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Macrophages/immunology , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Satellite Cells, Skeletal Muscle/immunology , Animals , Diabetes Mellitus, Experimental/pathology , Fibrosis , Indoleacetic Acids/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/drug effects , Necrosis , Regeneration , Satellite Cells, Skeletal Muscle/drug effects
13.
Diabetes ; 60(7): 1964-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21593201

ABSTRACT

OBJECTIVE: Type 1 diabetes leads to impairments in growth, function, and regenerative capacity of skeletal muscle; however, the underlying mechanisms have not been clearly defined. RESEARCH DESIGN AND METHODS: With the use of Ins2(WT/C96Y) mice (model of adolescent-onset type 1 diabetes), muscle regeneration was characterized in terms of muscle mass, myofiber size (cross-sectional area), and protein expression. Blood plasma was analyzed for glucose, nonesterified fatty acids, insulin, and plasminogen activator inhibitor-1 (PAI-1). PAI-039, an effective inhibitor of PAI-1, was orally administered to determine if PAI-1 was attenuating muscle regeneration in Ins2(WT/C96Y) mice. RESULTS: Ins2(WT/C96Y) mice exposed to 1 or 8 weeks of untreated type 1 diabetes before chemically induced muscle injury display significant impairments in their regenerative capacity as demonstrated by decreased muscle mass, myofiber cross-sectional area, myogenin, and Myh3 expression. PAI-1, a physiologic inhibitor of the fibrinolytic system and primary contributor to other diabetes complications, was more than twofold increased within 2 weeks of diabetes onset and remained elevated throughout the experimental period. Consistent with increased circulating PAI-1, regenerating muscles of diabetic mice exhibited excessive collagen levels at 5 and 10 days postinjury with concomitant decreases in active urokinase plasminogen activator and matrix metalloproteinase-9. Pharmacologic inhibition of PAI-1 with orally administered PAI-039 rescued the early regenerative impairments in noninsulin-treated Ins2(WT/C96Y) mice. CONCLUSIONS: Taken together, these data illustrate that the pharmacologic inhibition of elevated PAI-1 restores the early impairments in skeletal muscle repair observed in type 1 diabetes and suggests that early interventional studies targeting PAI-1 may be warranted to ensure optimal growth and repair in adolescent diabetic skeletal muscle.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Indoleacetic Acids/pharmacology , Muscle, Skeletal/physiology , Regeneration/drug effects , Animals , Male , Mice , Muscle, Skeletal/drug effects , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Urokinase-Type Plasminogen Activator/metabolism
14.
PLoS One ; 5(11): e14032, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-21103335

ABSTRACT

This present study investigated the temporal effects of type 1 diabetes mellitus (T1DM) on adolescent skeletal muscle growth, morphology and contractile properties using a 90% partial pancreatecomy (Px) model of the disease. Four week-old male Sprague-Dawley rats were randomly assigned to Px (n = 25) or Sham (n = 24) surgery groups and euthanized at 4 or 8 weeks following an in situ assessment of muscle force production. Compared to Shams, Px were hyperglycemic (>15 mM) and displayed attenuated body mass gains by days 2 and 4, respectively (both P<0.05). Absolute maximal force production of the gastrocnemius plantaris soleus complex (GPS) was 30% and 50% lower in Px vs. Shams at 4 and 8 weeks, respectively (P<0.01). GP mass was 35% lower in Px vs Shams at 4 weeks (1.24±0.06 g vs. 1.93±0.03 g, P<0.05) and 45% lower at 8 weeks (1.57±0.12 vs. 2.80±0.06, P<0.05). GP fiber area was 15-20% lower in Px vs. Shams at 4 weeks in all fiber types. At 8 weeks, GP type I and II fiber areas were ∼25% and 40% less, respectively, in Px vs. Shams (group by fiber type interactions, P<0.05). Phosphorylation states of 4E-BP1 and S6K1 following leucine gavage increased 2.0- and 3.5-fold, respectively, in Shams but not in Px. Px rats also had impaired rates of muscle protein synthesis in the basal state and in response to gavage. Taken together, these data indicate that exposure of growing skeletal muscle to uncontrolled T1DM significantly impairs muscle growth and function largely as a result of impaired protein synthesis in type II fibers.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Muscle, Skeletal/physiopathology , Muscular Diseases/physiopathology , Pancreatectomy/methods , Adolescent , Analgesics/pharmacology , Animals , Diabetes Mellitus, Type 1/complications , Disease Models, Animal , Humans , Ketamine/pharmacology , Male , Muscle Fatigue/drug effects , Muscle Fatigue/physiology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Diseases/etiology , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction , Succinate Dehydrogenase/metabolism , TOR Serine-Threonine Kinases/metabolism , Ubiquitination , Xylazine/pharmacology
15.
PLoS One ; 4(10): e7293, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19806198

ABSTRACT

BACKGROUND: The effects of diet-induced obesity on skeletal muscle function are largely unknown, particularly as it relates to changes in oxidative metabolism and morphology. PRINCIPAL FINDINGS: Compared to control fed mice, mice fed a high fat diet (HFD; 60% kcal: fat) for 8 weeks displayed increased body mass and insulin resistance without overt fasting hyperglycemia (i.e. pre-diabetic). Histological analysis revealed a greater oxidative potential in the HFD gastrocnemius/plantaris (increased IIA, reduced IIB fiber-type percentages) and soleus (increased I, IIA cross-sectional areas) muscles, but no change in fiber type percentages in tibialis anterior muscles compared to controls. Intramyocellular lipid levels were significantly increased relative to control in HFD gastrocnemius/plantaris, but were similar to control values in the HFD soleus. Using a novel, single muscle fiber approach, impairments in complete palmitate and glucose oxidation (72.8+/-6.6% and 61.8+/-9.1% of control, respectively; p<0.05) with HFD were detected. These reductions were consistent with measures made using intact extensor digitorum longus and soleus muscles. Compared to controls, no difference in succinate dehydrogenase or citrate synthase enzyme activities were observed between groups in any muscle studied, however, short-chain fatty acyl CoA dehydrogenase (SCHAD) activity was elevated in the HFD soleus, but not tibialis anterior muscles. Despite these morphological and metabolic alterations, no significant difference in peak tetanic force or low-frequency fatigue rates were observed between groups. CONCLUSIONS: These findings indicate that HFD induces early adaptive responses that occur in a muscle-specific pattern, but are insufficient to prevent impairments in oxidative metabolism with continued high-fat feeding. Moreover, the morphological and metabolic changes which occur with 8 weeks of HFD do not significantly impact muscle contractile properties.


Subject(s)
Animal Feed , Dietary Fats/metabolism , Muscle Contraction , Muscle, Skeletal/metabolism , Obesity/metabolism , Oxygen/metabolism , Animal Nutrition Sciences , Animals , Blood Glucose/metabolism , Glucose/metabolism , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Time Factors
16.
Am J Physiol Cell Physiol ; 295(1): C203-12, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18463233

ABSTRACT

Adiponectin (Ad) is linked to various disease states and mediates antidiabetic and anti-inflammatory effects. While it was originally thought that Ad expression was limited to adipocytes, we demonstrate here that Ad is expressed in mouse skeletal muscles and within differentiated L6 myotubes, as assessed by RT-PCR, Western blot, and immunohistochemical analyses. Serial muscle sections stained for fiber type, lipid content, and Ad revealed that muscle fibers with elevated intramyocellular Ad expression were consistently type IIA and IID fibers with detectably higher intramyocellular lipid (IMCL) content. To determine the effect of Ad on muscle phenotype and function, we used an Ad-null [knockout (KO)] mouse model. Body mass increased significantly in 24-wk-old KO mice [+5.5 +/- 3% relative to wild-type mice (WT)], with no change in muscle mass observed. IMCL content was significantly increased (+75.1 +/- 25%), whereas epididymal fat mass, although elevated, was not different in the KO mice compared with WT (+35.1 +/- 23%; P = 0.16). Fiber-type composition was unaltered, although type IIB fiber area was increased in KO mice (+25.5 +/- 6%). In situ muscle stimulation revealed lower peak tetanic forces in KO mice relative to WT (-47.5 +/- 6%), with no change in low-frequency fatigue rates. These data demonstrate that the absence of Ad expression causes contractile dysfunction and phenotypical changes in skeletal muscle. Furthermore, we demonstrate that Ad is expressed in skeletal muscle and that its intramyocellular localization is associated with elevated IMCL, particularly in type IIA/D fibers.


Subject(s)
Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/physiology , Adiponectin/biosynthesis , Adiponectin/genetics , Animals , Cell Line , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Phenotype
17.
Am J Physiol Heart Circ Physiol ; 293(6): H3692-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17921326

ABSTRACT

An issue central to understanding the biological benefits associated with regular exercise training is to elucidate the intracellular mechanisms governing exercise-conferred cardioprotection. Heat shock proteins (HSPs), most notably the inducible 70-kDa HSP family member Hsp70, are believed to participate in the protection of the myocardium during cardiovascular stress. Following acute exercise, activation of PKA mediates the suppression of an intermediary protein kinase, ERK1/2, which phosphorylates and suppresses the activation of the heat shock transcription factor 1 (HSF1). However, following exercise training, ERK1/2 has been reported to regulate the transcriptional activation of several genes involved in cell growth and proliferation and has been shown to be associated with training-mediated myocardial hypertrophy. The present project examined the transcriptional activation of hsp70 gene expression in acutely exercised (60 min at 30 m/min) naïve sedentary and aerobically trained (8 wk, low intensity) male Sprague-Dawley rats. Following acute exercise stress, no significant differences were demonstrated in the expression of myocardial Hsp70 mRNA and activation of PKA between sedentary and trained animals. However, trained animals elicited expression of the hsp70 gene (P < 0.05) in the presence of elevated ERK1/2 activation. Given the association of ERK1/2 and the suppression of hsp70 gene expression following acute exercise in naïve sedentary rats, these results suggest that training results in adaptations that allow for the simultaneous initiation of both proliferative and protective responses. While it is unclear what factors are associated with this training-related shift, increases in HSF1 DNA binding affinity (P < 0.05) and posttranscriptional modifications of the Hsp70 transcript are suggested.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Myocardium/metabolism , Physical Exertion/physiology , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Transcriptional Activation , Adaptation, Physiological , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation , Epinephrine/blood , HSP70 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/enzymology , Rats , Rats, Sprague-Dawley , Time Factors , Transcription Factors/metabolism , Up-Regulation
18.
J Mol Cell Cardiol ; 41(5): 816-22, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16806261

ABSTRACT

Exercise induces the expression of the cardioprotective protein, Hsp70, through the activation of its transcription factor HSF1. Recently, we reported that administration of a protein kinase A (PKA) inhibitor suppressed exercise-induced hsp70 gene expression, suggesting a role for PKA in the regulation of HSF1 activation in vivo. While the mechanism by which PKA regulates HSF1 is unclear, studies in vitro have reported that HSF1 is phosphorylated on two serine residues by mitogen activated protein kinases (MAPKs); ERK1/2 (ser307) and JNK/SAPK (ser363). As PKA is a regulator of these protein kinases, the current study examined the role of PKA in their activation and subsequent regulation of exercise-induced hsp70 gene expression. Following treadmill-running exercise (60 min at 30 m/min; 2% grade), both ERK1/2 and JNK/SAPK demonstrated distinct phosphorylation profiles. Increased phosphorylation of ERK1/2 was observed immediately post-exercise, whereas JNK/SAPK phosphorylation was not significantly elevated until 30 min post-exercise. Administration of the PKA inhibitor (H89; 0.360 mg/kg) maintained ERK1/2 phosphorylation to at least 30 min post exercise (n = 5; P < 0.05) while JNK/SAPK phosphorylation was unaltered. Inhibition of this PKA-mediated increase in ERK1/2 phosphorylation through the simultaneous administration of an ERK1/2 inhibitor (UB-1026; 0.25 mg/kg) restored exercise-induced hsp70 mRNA levels in PKA-inhibited rats that previously demonstrated a suppressed response (P < 0.05). Given that ERK1/2 has been shown to be a negative regulator of HSF1 in vitro, these results suggest a role for ERK1/2 in the PKA-mediated regulation of HSF1 activation following exercise.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , HSP70 Heat-Shock Proteins/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 3/metabolism , Physical Conditioning, Animal , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Heat Shock Transcription Factors , MAP Kinase Kinase 4/metabolism , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL