Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
1.
Crit Care ; 27(1): 428, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37932834

ABSTRACT

BACKGROUND: Critically ill patients suffer from acute muscle wasting, which is associated with significant physical functional impairment. We describe data from nested muscle biopsy studies from two trials of functional electrical stimulation (FES) that did not shown improvements in physical function. METHODS: Primary cohort: single-centre randomized controlled trial. Additional healthy volunteer data from patients undergoing elective hip arthroplasty. Validation cohort: Four-centre randomized controlled trial. INTERVENTION: FES cycling for 60-90min/day. ANALYSES: Skeletal muscle mRNA expression of 223 genes underwent hierarchal clustering for targeted analysis and validation. RESULTS: Positively enriched pathways between healthy volunteers and ICU participants were "stress response", "response to stimuli" and "protein metabolism", in keeping with published data. Positively enriched pathways between admission and day 7 ICU participants were "FOXO-mediated transcription" (admission = 0.48 ± 0.94, day 7 = - 0.47 ± 1.04 mean log2 fold change; P = 0.042), "Fatty acid metabolism" (admission = 0.50 ± 0.67, day 7 = 0.07 ± 1.65 mean log2 fold change; P = 0.042) and "Interleukin-1 processing" (admission = 0.88 ± 0.50, day 7 = 0.97 ± 0.76 mean log2 fold change; P = 0.054). Muscle mRNA expression of UCP3 (P = 0.030) and DGKD (P = 0.040) decreased in both cohorts with no between group differences. Changes in IL-18 were not observed in the validation cohort (P = 0.268). Targeted analyses related to intramuscular mitochondrial substrate oxidation, fatty acid oxidation and intramuscular inflammation showed PPARγ-C1α; (P < 0.001), SLC25A20 (P = 0.017) and UCP3 (P < 0.001) decreased between admission and day 7 in both arms. LPIN-1 (P < 0.001) and SPT1 (P = 0.044) decreased between admission and day 7. IL-18 (P = 0.011) and TNFRSF12A (P = 0.009) increased in both arms between admission and day 7. IL-1ß (P = 0.007), its receptor IL-1R1 (P = 0.005) and IL-6R (P = 0.001) decreased in both arms between admission and day 7. No between group differences were seen in any of these (all p > 0.05). CONCLUSIONS: Intramuscular inflammation and altered substrate utilization are persistent in skeletal muscle during first week of critical illness and are not improved by the application of Functional Electrical Stimulation-assisted exercise. Future trials of exercise to prevent muscle wasting and physical impairment are unlikely to be successful unless these processes are addressed by other means than exercise alone.


Subject(s)
Critical Illness , Interleukin-18 , Humans , Intensive Care Units , Muscular Atrophy , Electric Stimulation , Fatty Acids , RNA, Messenger , Membrane Transport Proteins
2.
J Nutr Health Aging ; 26(6): 637-651, 2022.
Article in English | MEDLINE | ID: mdl-35718874

ABSTRACT

Sarcopenia and frailty are highly prevalent conditions in older hospitalized patients, which are associated with a myriad of adverse clinical outcomes. This paper, prepared by a multidisciplinary expert working group from the Australian and New Zealand Society for Sarcopenia and Frailty Research (ANZSSFR), provides an up-to-date overview of current evidence and recommendations based on a narrative review of the literature for the screening, diagnosis, and management of sarcopenia and frailty in older patients within the hospital setting. It also includes suggestions on potential pathways to implement change to encourage widespread adoption of these evidence-informed recommendations within hospital settings. The expert working group concluded there was insufficient evidence to support any specific screening tool for sarcopenia and recommends an assessment of probable sarcopenia/sarcopenia using established criteria for all older (≥65 years) hospitalized patients or in younger patients with conditions (e.g., comorbidities) that may increase their risk of sarcopenia. Diagnosis of probable sarcopenia should be based on an assessment of low muscle strength (grip strength or five times sit-to-stand) with sarcopenia diagnosis including low muscle mass quantified from dual energy X-ray absorptiometry, bioelectrical impedance analysis or in the absence of diagnostic devices, calf circumference as a proxy measure. Severe sarcopenia is represented by the addition of impaired physical performance (slow gait speed). All patients with probable sarcopenia or sarcopenia should be investigated for causes (e.g., chronic/acute disease or malnutrition), and treated accordingly. For frailty, it is recommended that all hospitalized patients aged 70 years and older be screened using a validated tool [Clinical Frailty Scale (CFS), Hospital Frailty Risk Score, the FRAIL scale or the Frailty Index]. Patients screened as positive for frailty should undergo further clinical assessment using the Frailty Phenotype, Frailty Index or information collected from a Comprehensive Geriatric Assessment (CGA). All patients identified as frail should receive follow up by a health practitioner(s) for an individualized care plan. To treat older hospitalized patients with probable sarcopenia, sarcopenia, or frailty, it is recommended that a structured and supervised multi-component exercise program incorporating elements of resistance (muscle strengthening), challenging balance, and functional mobility training be prescribed as early as possible combined with nutritional support to optimize energy and protein intake and correct any deficiencies. There is insufficient evidence to recommend pharmacological agents for the treatment of sarcopenia or frailty. Finally, to facilitate integration of these recommendations into hospital settings organization-wide approaches are needed, with the Spread and Sustain framework recommended to facilitate organizational culture change, with the help of 'champions' to drive these changes. A multidisciplinary team approach incorporating awareness and education initiatives for healthcare professionals is recommended to ensure that screening, diagnosis and management approaches for sarcopenia and frailty are embedded and sustained within hospital settings. Finally, patients and caregivers' education should be integrated into the care pathway to facilitate adherence to prescribed management approaches for sarcopenia and frailty.


Subject(s)
Frailty , Sarcopenia , Aged , Aged, 80 and over , Australia , Frail Elderly , Frailty/diagnosis , Frailty/therapy , Geriatric Assessment , Hand Strength/physiology , Humans , New Zealand , Sarcopenia/diagnosis , Sarcopenia/therapy
3.
Transl Psychiatry ; 9(1): 166, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182707

ABSTRACT

Benefits of distributed learning strategies have been extensively described in the human literature, but minimally investigated in intellectual disability syndromes. We tested the hypothesis that training trials spaced apart in time could improve learning in two distinct genetic mouse models of neurodevelopmental disorders characterized by intellectual impairments. As compared to training with massed trials, spaced training significantly improved learning in both the Ts65Dn trisomy mouse model of Down syndrome and the maternally inherited Ube3a mutant mouse model of Angelman syndrome. Spacing the training trials at 1 h intervals accelerated acquisition of three cognitive tasks by Ts65Dn mice: (1) object location memory, (2) novel object recognition, (3) water maze spatial learning. Further, (4) spaced training improved water maze spatial learning by Ube3a mice. In contrast, (5) cerebellar-mediated rotarod motor learning was not improved by spaced training. Corroborations in three assays, conducted in two model systems, replicated within and across two laboratories, confirm the strength of the findings. Our results indicate strong translational relevance of a behavioral intervention strategy for improving the standard of care in treating the learning difficulties that are characteristic and clinically intractable features of many neurodevelopmental disorders.


Subject(s)
Behavior, Animal/physiology , Cognitive Remediation , Intellectual Disability/rehabilitation , Practice, Psychological , Recognition, Psychology/physiology , Spatial Learning/physiology , Spatial Memory/physiology , Angelman Syndrome/rehabilitation , Animals , Disease Models, Animal , Down Syndrome/rehabilitation , Female , Male , Mice , Mice, Knockout , Trisomy , Ubiquitin-Protein Ligases
4.
Cytokine ; 96: 274-278, 2017 08.
Article in English | MEDLINE | ID: mdl-28554144

ABSTRACT

Excessive inflammation reduces skeletal muscle protein synthesis leading to wasting and weakness. The janus kinase/signal transducers and activators of transcription-3 (JAK/STAT3) pathway is important for the regulation of inflammatory signaling. As such, suppressor of cytokine signaling-3 (SOCS3), the negative regulator of JAK/STAT signaling, is thought to be important in the control of muscle homeostasis. We hypothesized that muscle-specific deletion of SOCS3 would impair the anabolic response to leucine during an inflammatory insult. Twelve week old (n=8 per group) SOCS3 muscle-specific knockout mice (SOCS3-MKO) and littermate controls (WT) were injected with lipopolysaccharide (LPS, 1mg/kg) or saline and were studied during fasted conditions or after receiving 0.5g/kg leucine 3h after the injection of LPS. Markers of inflammation, anabolic signaling, and protein synthesis were measured 4h after LPS injection. LPS injection robustly increased mRNA expression of inflammatory molecules (Socs3, Socs1, Il-6, Ccl2, Tnfα and Cd68). In muscles from SOCS3-MKO mice, the Socs3 mRNA response to LPS was significantly blunted (∼6-fold) while STAT3 Tyr705 phosphorylation was exacerbated (18-fold). Leucine administration increased protein synthesis in both WT (∼1.6-fold) and SOCS3-MKO mice (∼1.5-fold) compared to basal levels. LPS administration blunted this effect, but there were no differences between WT and SOCS3-MKO mice. Muscle-specific SOCS3 deletion did not alter the response of AKT, mTOR, S6 or 4EBP1 under any treatment conditions. Therefore, SOCS3 does not appear to mediate the early inflammatory or leucine-induced changes in protein synthesis in skeletal muscle.


Subject(s)
Anabolic Agents , Inflammation/metabolism , Leucine/administration & dosage , Muscle, Skeletal/metabolism , Protein Biosynthesis , Suppressor of Cytokine Signaling 3 Protein/physiology , Animals , Chemokine CCL2/genetics , Disease Models, Animal , Interleukin-6/genetics , Leucine/metabolism , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Knockout , Phosphorylation , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics , Tumor Necrosis Factor-alpha/genetics
5.
J Physiol ; 587(Pt 7): 1593-605, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19204053

ABSTRACT

Excess lipid accumulation resulting from an elevated supply of plasma fatty acids is linked to the pathogenesis of the metabolic syndrome and heart disease. The term 'lipotoxicity' was coined to describe how lipid accumulation leads to cellular dysfunction and death in non-adipose tissues including the heart, pancreas and liver. While lipotoxicity has been shown in cultured skeletal muscle cells, the degree of lipotoxicity in vivo and the functional consequences are unresolved. We studied three models of fatty acid overload in male mice: 5 h Intralipid((R)) and heparin infusion, prolonged high fat feeding (HFF) and genetic obesity induced by leptin deficiency (ob/ob mice). Markers of apoptosis, proteolysis and autophagy were assessed as readouts of lipotoxicity. The Intralipid((R)) infusion increased caspase 3 activity in skeletal muscle, demonstrating that enhancing fatty acid flux activates pro-apoptotic pathways. HFF and genetic obesity increased tissue lipid content but did not influence apoptosis. Gene array analysis revealed that HFF reduced the expression of 31 pro-apoptotic genes. Markers of autophagy (LC3beta and beclin-1 expression) were unaffected by HFF and were associated with enhanced Bcl(2) protein expression. Proteolytic activity was similarly unaffected by HFF or in ob/ob mice. Thus, contrary to our previous findings in muscle culture in vitro and in other non-adipose tissues in vivo, lipid overload did not induce apoptosis, autophagy or proteolysis in skeletal muscle. A broad transcriptional suppression of pro-apoptotic proteins may explain this resistance to lipid-induced cell death in skeletal muscle.


Subject(s)
Dietary Fats/metabolism , Fatty Acids, Nonesterified/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Caspase 3/metabolism , Dietary Fats/administration & dosage , Disease Models, Animal , Down-Regulation , Fat Emulsions, Intravenous/metabolism , Fatty Acids, Nonesterified/blood , Gene Expression Profiling/methods , Hypertrophy , Leptin/deficiency , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Obesity/genetics , Obesity/pathology , Oligonucleotide Array Sequence Analysis , Proteasome Endopeptidase Complex/metabolism , Time Factors , Transcription, Genetic
6.
Int J Biochem Cell Biol ; 39(3): 469-77, 2007.
Article in English | MEDLINE | ID: mdl-17137828

ABSTRACT

Duchenne muscular dystrophy is a lethal X-linked muscle disease resulting from a defect in the muscle membrane protein dystrophin. The absence of dystrophin leads to muscle membrane fragility, muscle death (necrosis) and eventual replacement of skeletal muscle by fat and fibrous connective tissue. Extensive muscle wasting and respiratory failure results in premature death often by the early 20s. This short review evaluates drug and nutritional interventions designed to reduce the severity of muscular dystrophy, while awaiting the outcome of research into therapies to correct the fundamental gene defect. Combinations of dietary supplementation with amino-acids such as creatine, specific anti-inflammatory drugs and perhaps drugs that target ion channels might have immediate realistic clinical benefits although rigorous research is required to determine optimal combinations of such interventions.


Subject(s)
Muscular Dystrophy, Duchenne/diet therapy , Muscular Dystrophy, Duchenne/drug therapy , Adrenal Cortex Hormones/therapeutic use , Adrenergic beta-Agonists/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Cytokines/antagonists & inhibitors , Dietary Supplements , Humans , Ion Channels/metabolism , Mice , Mice, Inbred mdx , Muscular Dystrophy, Animal/diet therapy , Muscular Dystrophy, Animal/drug therapy , Protease Inhibitors/therapeutic use
7.
Gene Ther ; 13(23): 1657-64, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16871234

ABSTRACT

Developing methodologies to enhance skeletal muscle regeneration and hasten the restoration of muscle function has important implications for minimizing disability after injury and for treating muscle diseases such as Duchenne muscular dystrophy. Although delivery of various growth factors, such as insulin-like growth factor-I (IGF-I), have proved successful in promoting skeletal muscle regeneration after injury, no study has compared the efficacy of different delivery methods directly. We compared the efficacy of systemic delivery of recombinant IGF-I protein via mini-osmotic pump (approximately 1.5 mg/kg/day) with a single electrotransfer-assisted plasmid-based gene transfer, to hasten functional repair of mouse tibialis anterior muscles after myotoxic injury. The relative efficacy of each method was assessed at 7, 21 and 28 days post-injury. Our findings indicate that IGF-I hastened functional recovery, regardless of the route of IGF-I administration. However, gene transfer of IGF-I was superior to systemic protein administration because in the regenerating muscle, this delivery method increased IGF-I levels, activated intracellular signals (Akt phosphorylation), induced a greater magnitude of myofiber hypertrophy and hastened functional recovery at an earlier time point (14 days) after injury than did protein administration (21 days). Thus, the relative efficacy of different modes of delivery is an important consideration when assessing the therapeutic potential of various proteins for treating muscle injuries and skeletal muscle diseases.


Subject(s)
Genetic Therapy/methods , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/genetics , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Regeneration , Animals , DNA/administration & dosage , Drug Implants , Electroporation/methods , Gene Expression , Immunohistochemistry/methods , Injections, Intramuscular , Insulin-Like Growth Factor I/analysis , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Muscle Contraction , Muscle, Skeletal/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Time Factors
8.
Intern Med J ; 34(5): 294-6, 2004 May.
Article in English | MEDLINE | ID: mdl-15151679

ABSTRACT

Some of the most serious consequences of ageing are its effects on skeletal muscle structure and function. Ageing is associated with a progressive loss of muscle mass (sarcopenia), a slowing of movement, and a decline in strength; factors that increase the risk of injury from sudden falls and a reliance on the frail elderly for assistance in accomplishing even the most basic tasks required for independent living. From a public health perspective, sarcopenia has widespread clinical implications. As the proportion of older persons in the population continues to grow, sarcopenia will have a dramatic impact on the lives of Australians and place increasing demands on health care. Although it is generally agreed that the deleterious effects of ageing on skeletal muscle are inevitable, debate exists as to whether these intrinsic changes are immutable or reversible. There is clearly a profound need for therapeutic strategies that can slow the effects of ageing on muscle function, and restore muscle size and strength in the frail elderly so that their quality of life can be maintained or improved. Physical activity plays an important role in slowing the effects of ageing, but exercise alone will not prevent the gradual decline in skeletal muscle function. Other factors, such as age-related changes in circulating levels of muscle anabolic hormones and growth factors, must also be considered when developing strategies to combat sarcopenia. Much research is needed to test the safety and efficacy of these exciting experimental strategies before they can be recommended for clinical application.


Subject(s)
Aging , Muscle Weakness/physiopathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology , Activities of Daily Living , Australia/epidemiology , Exercise , Humans , Motor Activity , Muscle Weakness/drug therapy , Muscle Weakness/epidemiology , Muscle Weakness/prevention & control , Muscular Atrophy/drug therapy , Muscular Atrophy/epidemiology , Muscular Atrophy/prevention & control , Population Dynamics
9.
Exerc Sport Sci Rev ; 29(4): 141-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11688785

ABSTRACT

Muscle atrophy or wasting is a loss of muscle tissue resulting from disease or lack of use. This review examines recent pharmacologic or nutrition interventions for ameliorating wasting and improving muscle function in neuromuscular disorders. The information has application for treating the muscular dystrophies, cancer cachexia, weightlessness, immobilization, denervation, and disuse atrophy.


Subject(s)
Muscular Dystrophy, Duchenne/therapy , Neuromuscular Diseases/therapy , Adjuvants, Immunologic/therapeutic use , Adrenergic beta-Agonists/therapeutic use , Albuterol/therapeutic use , Aminoglycosides , Animals , Anti-Bacterial Agents/therapeutic use , Creatine/therapeutic use , Cytokines/therapeutic use , Dehydroepiandrosterone/therapeutic use , Glucocorticoids/therapeutic use , Glutamine/therapeutic use , Human Growth Hormone/therapeutic use , Humans , Insulin-Like Growth Factor I/therapeutic use , Muscle, Skeletal/drug effects , Neuromuscular Diseases/physiopathology , Testosterone/therapeutic use , Valerates
10.
Clin Exp Pharmacol Physiol ; 28(9): 779-81, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11553038

ABSTRACT

1. Ageing is generally associated with a decline in skeletal muscle mass and strength and a slowing of muscle contraction, factors that impact upon the quality of life for the elderly. The mechanisms underlying this age-related muscle weakness have not been fully resolved. The purpose of the present study was to determine whether the decrease in muscle force as a consequence of age could be attributed partly to a decrease in the number of cross-bridges participating during contraction. 2. Given that the rigor force is proportional to the approximate total number of interacting sites between the actin and myosin filaments, we tested the null hypothesis that the rigor force of permeabilized muscle fibres from young and old rats would not be different. 3. Permeabilized fibres from the extensor digitorum longus (fast-twitch; EDL) and soleus (predominantly slow-twitch) muscles of young (6 months of age) and old (27 months of age) male F344 rats were activated in Ca2+-buffered solutions to determine force-pCa characteristics (where pCa = -log(10)[Ca2+]) and then in solutions lacking ATP and Ca2+ to determine rigor force levels. 4. The rigor forces for EDL and soleus muscle fibres were not different between young and old rats, indicating that the approximate total number of cross-bridges that can be formed between filaments did not decline with age. We conclude that the age-related decrease in force output is more likely attributed to a decrease in the force per cross-bridge and/or decreases in the efficiency of excitation-contraction coupling.


Subject(s)
Aging/physiology , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/physiology , Animals , Calcium/pharmacology , Cell Membrane Permeability , Male , Muscle Contraction/drug effects , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Muscle, Skeletal/drug effects , Rats , Rats, Inbred F344 , Strontium/pharmacology
11.
J Physiol ; 535(Pt 2): 591-600, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11533147

ABSTRACT

1. Differences in the effect of age on structure-function relationships of limb muscles of mdx (dystrophin null) and control mice have not been resolved. We tested the hypotheses that, compared with limb muscles from age-matched control mice, limb muscles of 6- to 17-month-old mdx mice are larger but weaker, with lower normalised force and power, whereas those from 24- to 28-month-old mdx mice are smaller and weaker. 2. The maximum isometric tetanic force (P(o)) and power output of limb muscles from 6-, 17-, 24- and 28-month-old mdx and control mice were measured in vitro at 25 degrees C and normalised with respect to cross-sectional area and muscle mass, respectively. 3. Body mass at 6 and 28 months was not significantly different in mdx and control mice, but that of control mice increased 16 % by 17 months and then declined 32 % by 28 months. The body masses of mdx mice declined linearly with age with a decrease of 25 % by 28 months. From 6 to 28 months of age, the range in the decline in the masses of EDL and soleus muscles of mdx and control mice was from 16 to 28 %. The muscle masses of mdx mice ranged from 9 % to 42 % greater than those of control mice at each of the four ages and, even at 28 months, the masses of EDL and soleus muscles of mdx mice were 17 % and 22 % greater than control values. 4. For mdx mice of all ages, muscle hypertrophy was highly effective in the maintenance of control values for absolute force for both EDL and soleus muscles and for absolute power of soleus muscles. Throughout their lifespan, muscles of mdx mice displayed significant weakness with values for specific P(o) and normalised power approximately 20 % lower than values for control mice at each age. For muscles of both strains, normalised force and power decreased approximately 28 % with age, and consequently weakness was more severe in muscles of old mdx than in those of old control mice.


Subject(s)
Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscular Dystrophy, Animal/physiopathology , Animals , Dystrophin/genetics , Isometric Contraction/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Organ Size , Specific Pathogen-Free Organisms
12.
Hum Gene Ther ; 12(6): 629-42, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11426463

ABSTRACT

Targeted genetic correction of mutations in cells is a potential strategy for treating human conditions that involve nonsense, missense, and transcriptional splice junction mutations. One method of targeted gene repair, single-stranded short-fragment homologous replacement (ssSFHR), has been successful in repairing the common deltaF508 3-bp microdeletion at the cystic fibrosis transmembrane conductance regulator (CFTR) locus in 1% of airway epithelial cells in culture. This study investigates in vitro and in vivo application of a double-stranded method variant of SFHR gene repair to the mdx mouse model of Duchenne muscular dystrophy (DMD). A 603-bp wild-type PCR product was used to repair the exon 23 C-to-T mdx nonsense transition at the Xp21.1 dys locus in cultured myoblasts and in tibialis anterior (TA) from male mdx mice. Multiple transfection and variation of lipofection reagent both improved in vitro SFHR efficiency, with successful conversion of mdx to wild-type nucleotide at the dys locus achieved in 15 to 20% of cultured loci and in 0.0005 to 0.1% of TA. The genetic correction of mdx myoblasts was shown to persist for up to 28 days in culture and for at least 3 weeks in TA. While a high frequency of in vitro gene repair was observed, the lipofection used here appeared to have adverse effects on subsequent cell viability and corrected cells did not express dystrophin transcript. With further improvements to in vitro and in vivo gene repair efficiencies, SFHR may find some application in DMD and other genetic neuromuscular disorders in humans.


Subject(s)
Codon, Nonsense , DNA Repair/genetics , Dystrophin/genetics , Genetic Therapy/methods , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/therapy , Animals , Cation Exchange Resins , Cell Transplantation , Dystrophin/deficiency , Dystrophin/metabolism , Female , Gene Deletion , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Immunoenzyme Techniques , In Vitro Techniques , Lipids , Liposomes , Male , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscular Dystrophy, Animal/genetics , Polymorphism, Restriction Fragment Length , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transfection/methods
13.
Neuromuscul Disord ; 11(3): 260-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11297941

ABSTRACT

Although insulin-like growth factor-I (IGF-I) has been proposed for use by patients suffering from muscle wasting conditions, few studies have investigated the functional properties of dystrophic skeletal muscle following IGF-I treatment. 129P1 ReJ-Lama2(dy) (129 ReJ dy/dy) dystrophic mice suffer from a deficiency in the structural protein, laminin, and exhibit severe muscle wasting and weakness. We tested the hypothesis that 4 weeks of IGF-I treatment ( approximately 2 mg/kg body mass, 50 g/h via mini-osmotic pump, subcutaneously) would increase the mass and force producing capacity of skeletal muscles from dystrophic mice. IGF-I treatment increased the mass of the extensor digitorum longus (EDL) and soleus muscles of dystrophic mice by 20 and 29%, respectively, compared with untreated dystrophic mice (administered saline-vehicle only). Absolute maximum force (P(o)) of the EDL and soleus muscle was increased by 40 and 32%, respectively, following IGF-I treatment. Specific P(o) (sP(o)) was increased by 23% in the EDL muscles of treated compared with untreated mice, but in the soleus muscle sP(o) was unchanged. IGF-I treatment increased the proportion of type IIB and type IIA fibres and decreased the proportion of type I fibres in the EDL muscles of dystrophic mice. In the soleus muscles of dystrophic mice, IGF-I treatment increased the proportion of type IIA fibres and decreased the proportion of type I fibres. Average fibre cross-sectional area was increased in the EDL and soleus muscles of treated compared with untreated mice. We conclude that IGF-I treatment ameliorates muscle wasting and improves the functional properties of skeletal muscles of dystrophic mice. The findings have important implications for the role of IGF-I in ameliorating muscle wasting associated with the muscular dystrophies.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Muscle, Skeletal/drug effects , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Animals , Cell Size/drug effects , Cell Size/physiology , Disease Models, Animal , Male , Mice , Mice, Mutant Strains , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Organ Size/drug effects , Organ Size/physiology
14.
J Neurophysiol ; 85(3): 1027-32, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11247972

ABSTRACT

Extraocular muscles are characterized by their faster rates of contraction and their higher resistance to fatigue relative to limb skeletal muscles. Another often reported characteristic of extraocular muscles is that they generate lower specific forces (sP(o), force per muscle cross-sectional area, kN/m(2)) than limb skeletal muscles. To investigate this perplexing issue, the isometric contractile properties of the levator palpebrae superioris (levator) and superior rectus muscles of the rat were examined in situ with nerve and blood supply intact. The extraocular muscles were attached to a force transducer, and the cranial nerves exposed for direct stimulation. After determination of optimal muscle length (L(o)) and stimulation voltage, a full frequency-force relationship was established for each muscle. Maximum isometric tetanic force (P(o)) for the levator and superior rectus muscles was 177 +/- 13 and 280 +/- 10 mN (mean +/- SE), respectively. For the calculation of specific force, a number of rat levator and superior rectus muscles were stored in a 20% nitric acid-based solution to isolate individual muscle fibers. Muscle fiber lengths (L(f)) were expressed as a percentage of overall muscle length, allowing a mean L(f) to L(o) ratio to be used in the estimation of muscle cross-sectional area. Mean L(f):L(o) was determined to be 0.38 for the levator muscle and 0.45 for the superior rectus muscle. The sP(o) for the rat levator and superior rectus muscles measured in situ was 275 and 280 kN/m(2), respectively. These values are within the range of sP(o) values commonly reported for rat skeletal muscles. Furthermore P(o) and sP(o) for the rat levator and superior rectus muscles measured in situ were significantly higher (P < 0.001) than P(o) and sP(o) for these muscles measured in vitro. The results indicate that the force output of intact extraocular muscles differs greatly depending on the mode of testing. Although in vitro evaluation of extraocular muscle contractility will continue to reveal important information about this group of understudied muscles, the lower sP(o) values of these preparations should be recognized as being significantly less than their true potential. We conclude that extraocular muscles are not intrinsically weaker than skeletal muscles.


Subject(s)
Isometric Contraction/physiology , Oculomotor Muscles/physiology , Animals , Cerebral Decortication , Electric Stimulation , Female , In Vitro Techniques , Male , Muscle Fibers, Skeletal/physiology , Oculomotor Muscles/blood supply , Oculomotor Muscles/innervation , Rats , Rats, Sprague-Dawley , Stress, Mechanical
15.
Neuromuscul Disord ; 11(2): 192-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11257477

ABSTRACT

Based on its anabolic properties, treatment with the beta(2)-adrenoceptor agonist, clenbuterol, has been proposed as a strategy for ameliorating the symptoms of muscular dystrophy. In the dystrophic mdx mouse, only the diaphragm muscle exhibits progressive and severe degeneration in muscle structure and function similar to that observed in Duchenne muscular dystrophy. We tested the hypothesis that 20 weeks of clenbuterol treatment ( approximately 1.5-2 mg kg(-1)day(-1)) would increase the force and power output of diaphragm muscle strips of 6-month-old mdx and control mice. At this age, the diaphragm muscles of mdx mice show extensive degeneration and impaired contractility compared with control mice. Clenbuterol treatment did not increase the normalized force or power output of diaphragm strips from either mdx or control mice. The degeneration and necrosis within the diaphragm muscle of mdx mice was also not ameliorated by clenbuterol treatment. The results indicate that clenbuterol treatment does not improve the structure or function of diaphragm muscles from mdx mice.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Clenbuterol/pharmacology , Diaphragm/drug effects , Disease Models, Animal , Muscle Contraction/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Animals , Diaphragm/pathology , Diaphragm/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction/physiology , Muscle Weakness/drug therapy , Muscle Weakness/pathology , Muscle Weakness/physiopathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology
16.
J Appl Physiol (1985) ; 90(3): 832-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181590

ABSTRACT

We used intact fast-twitch extensor digitorum longus (EDL) and slow-twitch soleus muscles from rats and mice to test the hypothesis that exogenous application of an oxidant would increase maximum isometric force production (P(o)) of slow-twitch muscles to a greater extent than fast-twitch skeletal muscles. Exposure to an oxidant, hydrogen peroxide (H(2)O(2); 100 microM to 5 mM, 30 min), affected P(o) of rat muscles in a time- and dose-dependent manner. P(o) of rat soleus muscles was increased by 8 +/- 1 (SE) and 14 +/- 1% (P < 0.01) after incubation with 1 and 5 mM H(2)O(2), respectively, whereas in mouse soleus muscles P(o) was only increased after incubation with 500 microM H(2)O(2). P(o) of rat EDL muscles was affected by H(2)O(2) biphasically; initially there was a small increase (3 +/- 1%), but then P(o) diminished significantly after 30 min of treatment. In contrast, all concentrations of H(2)O(2) tested decreased P(o) of mouse EDL muscles. A reductant, dithiothreitol (DTT; rat = 10 mM, mouse = 1 mM), was added to quench H(2)O(2), and it reversed the potentiation in P(o) in rat soleus but not in rat EDL muscles or in any H(2)O(2)-treated mouse muscles. After prolonged equilibration (30 min) with 5 mM H(2)O(2) without prior activation, P(o) was potentiated in rat soleus but not EDL muscles, demonstrating that the effect of oxidation in the soleus muscles was also dependent on the activation history of the muscle. The results of these experiments demonstrate that P(o) of both slow- and fast-twitch muscles from rats and mice is modified by redox modulation, indicating that maximum P(o) of mammalian skeletal muscles is dependent on oxidation.


Subject(s)
Isometric Contraction/physiology , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/physiology , Animals , Dithiothreitol/pharmacology , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Isometric Contraction/drug effects , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Muscle, Skeletal/drug effects , Organ Specificity , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
17.
Eur J Appl Physiol ; 86(1): 24-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11820317

ABSTRACT

In skeletal muscle the activity of the enzymatic antioxidants superoxide dismutase (SOD), glutathione peroxidase (GPx) and catalase (CAT) is regulated in response to generation of reactive oxygen species (ROS). Increased activity of these enzymes is observed after repeated bouts of aerobic exercise, a potent stimulus for intracellular ROS production. Hyperbaric oxygen (HBO) inhalation also stimulates intracellular ROS production although the effects of HBO on skeletal muscle SOD, GPx and CAT activity have not been studied. We tested the hypothesis that SOD, GPx and CAT activity is modulated in skeletal muscles in response to acute and repeated HBO administration. In adult male rats acute HBO inhalation (60 mm at 3 atmospheres absolute) reduced catalase activity by approximately 51% in slow-twitch soleus muscles. Additionally, repeated HBO inhalation (twice daily for 28 days) increased Mn2+-superoxide dismutase activity by approximately 241% in fast-twitch extensor digitorum longus muscles. We conclude that both acute and repeated HBO inhalation can alter enzymatic antioxidant activity in skeletal muscles.


Subject(s)
Hyperbaric Oxygenation , Muscle, Skeletal/enzymology , Oxidoreductases/metabolism , Oxygen/pharmacology , Animals , Antioxidants/metabolism , Catalase/metabolism , Glutathione Peroxidase/metabolism , Male , Muscle Fibers, Fast-Twitch/enzymology , Muscle Fibers, Slow-Twitch/enzymology , Muscle, Skeletal/cytology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
18.
Muscle Nerve ; 23(11): 1700-5, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11054748

ABSTRACT

Although the muscles of the mdx mouse lack dystrophin, the protein absent in muscles of humans affected with Duchenne muscular dystrophy (DMD), the only mdx muscle to degenerate in a manner similar to those of DMD boys is the diaphragm. We have previously shown that leukemia inhibitory factor (LIF) is a trauma factor that enhances muscle repair in vivo and, when applied exogenously, increases the fiber size of mdx skeletal muscle. Furthermore, we developed a controlled release device for LIF based on a calcium alginate rod (release rate about 0.5% per day). These rods were sutured to the abdominal surface of the hemidiaphragm of mdx mice 3 months old. At age 6 months the mice were killed and the diaphragm muscles fixed and sectioned. The sections showed obvious muscle degeneration at 3 months of age in mdx mouse diaphragms and further degeneration at 6 months in saline-perfused muscle. Hemidiaphragm muscles continuously exposed to LIF over the same period contained more normal myofibers, larger regenerated fibers, and less adipose tissue and other non-contractile tissue. Morphometric analysis of the diaphragm sections was carried out. The LIF-treated animals showed a significant increase in fiber number and size compared to saline rod controls. The amount of nonmuscle (connective tissue and adipose tissue) was significantly reduced and the maximum force-producing capacity of isolated diaphragm muscle strips was higher in LIF-treated mice. The results demonstrate that LIF treatment ameliorates the dystrophic abnormalities in mdx mouse diaphragm.


Subject(s)
Growth Inhibitors/pharmacology , Interleukin-6 , Lymphokines/pharmacology , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/drug therapy , Muscular Atrophy/pathology , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Animal/pathology , Animals , Cell Size/drug effects , Diaphragm/cytology , Diaphragm/pathology , Diaphragm/physiology , Infusion Pumps, Implantable , Leukemia Inhibitory Factor , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle Fatigue/drug effects , Recombinant Proteins/pharmacology
19.
Am J Physiol Cell Physiol ; 279(4): C1290-4, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003610

ABSTRACT

Muscle fibers of mdx mice that lack dystrophin are more susceptible to contraction-induced injury, particularly when stretched. In contrast, transgenic mdx (tg-mdx) mice, which overexpress dystrophin, show no morphological or functional signs of dystrophy. Permeabilization disrupts the sarcolemma of fibers from muscles of mdx, tg-mdx, and control mice. We tested the null hypothesis stating that, after single stretches of maximally activated single permeabilized fibers, force deficits do not differ among fibers from extensor digitorum longus muscles of mdx, tg-mdx, or control mice. Fibers were maximally activated by Ca(2+) (pCa 4.5) and then stretched through strains of 10%, 20%, or 30% of fiber length (L(f)) at a velocity of 0.5 L(f)/s. Immediately after each strain, the force deficits were not different for fibers from each of the three groups of mice. When collated with studies of membrane-intact fibers in whole muscles of mdx, tg-mdx, and control mice, these results indicate that dystrophic symptoms do not arise from factors within myofibrils but, rather, from disruption of the sarcolemmal integrity that normally provides protection from contraction-induced injury.


Subject(s)
Cell Membrane Permeability/genetics , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/physiopathology , Animals , Calcium/metabolism , Calcium/pharmacology , Dystrophin/metabolism , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Transgenic , Muscle Contraction/genetics , Muscle Fibers, Skeletal/drug effects , Sarcolemma/genetics , Sarcolemma/metabolism , Stress, Mechanical
20.
J Appl Physiol (1985) ; 89(4): 1477-82, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11007585

ABSTRACT

There is growing interest in hyperbaric oxygen (HBO) as an adjunctive treatment for muscle injuries. This experiment tested the hypothesis that periodic inhalation of HBO hastens the functional recovery and myofiber regeneration of skeletal muscle after myotoxic injury. Injection of the rat extensor digitorum longus (EDL) muscle with bupivacaine hydrochloride causes muscle degeneration. After injection, rats breathed air with or without periodic HBO [100% O(2) at either 2 or 3 atmospheres absolute (ATA)]. In vitro maximum isometric tetanic force of injured EDL muscles and regenerating myofiber size were unchanged between 2 ATA HBO-treated and untreated rats at 14 days postinjury but were approximately 11 and approximately 19% greater, respectively, in HBO-treated rats at 25 days postinjury. Maximum isometric tetanic force of injured muscles was approximately 27% greater, and regenerating myofibers were approximately 41% larger, in 3 ATA HBO-treated rats compared with untreated rats at 14 days postinjury. These findings demonstrate that periodic HBO inhalation increases maximum force-producing capacity and enhances myofiber growth in regenerating skeletal muscle after myotoxic injury with greater effect at 3 than at 2 ATA.


Subject(s)
Hyperbaric Oxygenation , Isometric Contraction/physiology , Muscle, Skeletal/physiology , Regeneration/physiology , Animals , Bupivacaine/toxicity , Male , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...