Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 19(11)2018 Nov 12.
Article in English | MEDLINE | ID: mdl-30424476

ABSTRACT

Molecular mechanisms that trigger disuse-induced postural muscle atrophy as well as myosin phenotype transformations are poorly studied. This review will summarize the impact of 5' adenosine monophosphate -activated protein kinase (AMPK) activity on mammalian target of rapamycin complex 1 (mTORC1)-signaling, nuclear-cytoplasmic traffic of class IIa histone deacetylases (HDAC), and myosin heavy chain gene expression in mammalian postural muscles (mainly, soleus muscle) under disuse conditions, i.e., withdrawal of weight-bearing from ankle extensors. Based on the current literature and the authors' own experimental data, the present review points out that AMPK plays a key role in the regulation of signaling pathways that determine metabolic, structural, and functional alternations in skeletal muscle fibers under disuse.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Atrophy/enzymology , Muscular Disorders, Atrophic/enzymology , Animals , Energy Metabolism , Humans , Muscular Atrophy/pathology , Muscular Disorders, Atrophic/pathology , Signal Transduction
2.
J Cell Physiol ; 231(5): 1163-70, 2016 May.
Article in English | MEDLINE | ID: mdl-26460818

ABSTRACT

Unloading induces bone loss and causes disuse osteoporosis. However, the mechanism underlying disuse osteoporosis is still incompletely understood. Here, we examined the effects of cathepsin K (CatK) deficiency on disuse osteoporosis induced by using sciatic neurectomy (Nx) model. After 4 weeks of surgery, CatK KO and WT mice were sacrificed and subjected to analyses. For cancellous bone rich region, Nx reduced the bone mineral density (BMD) compared to the BMD in the sham operated side in wild type mice. In contrast, CatK deficiency suppressed such Nx-induced reduction of BMD in cancellous bone. Nx also reduced BMD in the mid shaft cortical bone compared to the BMD in the corresponding region on the sham operated side in wild type mice. In contrast, CatK deficiency suppressed such Nx-induced reduction of BMD in the mid shaft cortical bone. Bone volume (BV/TV) was reduced by Nx in WT mice. In contrast, Cat-K deficiency suppressed such reduction in bone volume. Interestingly, CatK deficiency suppressed osteoclast number and osteoclast surface in the Nx side compared to sham side. When bone marrow cells obtained from Nx side femur of CatK-KO mice were cultured, the levels of the calcified area in culture were increased. Further examination of gene expression indicated that Nx suppressed the expression of genes encoding osteoblast-phenotype-related molecules such as Runx2 and alkaline phosphatase in WT mice. In contrast, CatK deficiency suppressed such reduction. These data indicate that CatK is involved in the disuse-induced bone mass reduction.


Subject(s)
Bone Resorption/enzymology , Bone Resorption/etiology , Cathepsin K/deficiency , Muscular Disorders, Atrophic/complications , Muscular Disorders, Atrophic/enzymology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Density , Bone Marrow Cells/metabolism , Bone Resorption/diagnostic imaging , Bone Resorption/physiopathology , Bone and Bones/diagnostic imaging , Bone and Bones/enzymology , Bone and Bones/pathology , Calcification, Physiologic/genetics , Cathepsin K/metabolism , Cells, Cultured , Imaging, Three-Dimensional , Mice, Inbred C57BL , Muscular Disorders, Atrophic/diagnostic imaging , Muscular Disorders, Atrophic/pathology , Organ Size , Osteoclasts/pathology , Osteogenesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , X-Ray Microtomography
3.
Biomed Res Int ; 2015: 720172, 2015.
Article in English | MEDLINE | ID: mdl-25654120

ABSTRACT

This study examines the isoform-specific effects of short-term hindlimb suspension (HS) on the Na,K-ATPase in rat soleus muscle. Rats were exposed to 24-72 h of HS and we analyzed the consequences on soleus muscle mass and contractile parameters; excitability and the resting membrane potential (RMP) of muscle fibers; the electrogenic activity, protein, and mRNA content of the α1 and α2 Na,K-ATPase; the functional activity and plasma membrane localization of the α2 Na,K-ATPase. Our results indicate that 24-72 h of HS specifically decreases the electrogenic activity of the Na,K-ATPase α2 isozyme and the RMP of soleus muscle fibers. This decrease occurs prior to muscle atrophy or any change in contractile parameters. The α2 mRNA and protein content increased after 24 h of HS and returned to initial levels at 72 h; however, even the increased content was not able to restore α2 enzyme activity in the disused soleus muscle. There was no change in the membrane localization of α2 Na,K-ATPase. The α1 Na,K-ATPase electrogenic activity, protein and mRNA content did not change. Our findings suggest that skeletal muscle use is absolutely required for α2 Na,K-ATPase transport activity and provide the first evidence that Na,K-ATPase alterations precede HS-induced muscle atrophy.


Subject(s)
Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Disorders, Atrophic/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Body Weight , Hindlimb Suspension , Isoenzymes/metabolism , Male , Membrane Potentials , Muscle Contraction , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Muscular Disorders, Atrophic/pathology , Muscular Disorders, Atrophic/physiopathology , Nicotine/pharmacology , Organ Size , Rats, Wistar
4.
Neuron ; 85(1): 88-100, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25569348

ABSTRACT

Polyglutamine expansion in androgen receptor (AR) is responsible for spinobulbar muscular atrophy (SBMA) that leads to selective loss of lower motor neurons. Using SBMA as a model, we explored the relationship between protein structure/function and neurodegeneration in polyglutamine diseases. We show here that protein arginine methyltransferase 6 (PRMT6) is a specific co-activator of normal and mutant AR and that the interaction of PRMT6 with AR is significantly enhanced in the AR mutant. AR and PRMT6 interaction occurs through the PRMT6 steroid receptor interaction motif, LXXLL, and the AR activating function 2 surface. AR transactivation requires PRMT6 catalytic activity and involves methylation of arginine residues at Akt consensus site motifs, which is mutually exclusive with serine phosphorylation by Akt. The enhanced interaction of PRMT6 and mutant AR leads to neurodegeneration in cell and fly models of SBMA. These findings demonstrate a direct role of arginine methylation in polyglutamine disease pathogenesis.


Subject(s)
Drosophila Proteins/genetics , Muscular Disorders, Atrophic/enzymology , Peptides/genetics , Protein-Arginine N-Methyltransferases/metabolism , RNA, Messenger/analysis , Receptors, Androgen/metabolism , Animals , COS Cells , Chlorocebus aethiops , Drosophila , Drosophila Proteins/metabolism , HEK293 Cells , Humans , Mice , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/metabolism , Nuclear Proteins/metabolism , PC12 Cells , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics
5.
Antioxid Redox Signal ; 20(16): 2479-96, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24093939

ABSTRACT

AIMS: Redox and growth-factor imbalance fosters muscle disuse atrophy. Since the endoplasmic-reticulum chaperone Grp94 is required for folding insulin-like growth factors (IGFs) and for antioxidant cytoprotection, we investigated its involvement in muscle mass loss due to inactivity. RESULTS: Rat soleus muscles were transfected in vivo and analyzed after 7 days of hindlimb unloading, an experimental model of muscle disuse atrophy, or standard caging. Increased muscle protein carbonylation and decreased Grp94 protein levels (p<0.05) characterized atrophic unloaded solei. Recombinant Grp94 expression significantly reduced atrophy of transfected myofibers, compared with untransfected and empty-vector transfected ones (p<0.01), and decreased the percentage of carbonylated myofibers (p=0.001). Conversely, expression of two different N-terminal deleted Grp94 species did not attenuate myofiber atrophy. No change in myofiber trophism was detected in transfected ambulatory solei. The absence of effects on atrophic untransfected myofibers excluded a major role for IGFs folded by recombinant Grp94. Immunoprecipitation and confocal microscopy assays to investigate chaperone interaction with muscle atrophy regulators identified 160 kDa neuronal nitric oxide synthase (nNOS) as a new Grp94 partner. Unloading was demonstrated to untether nNOS from myofiber subsarcolemma; here, we show that such nNOS localization, revealed by means of NADPH-diaphorase histochemistry, appeared preserved in unloaded myofibers expressing recombinant Grp94, compared to those transfected with the empty vector or deleted Grp94 cDNA (p<0.02). INNOVATION: Grp94 interacts with nNOS and prevents its untethering from sarcolemma in unloaded myofibers. CONCLUSION: Maintenance of Grp94 expression is sufficient to counter unloading atrophy and oxidative stress by mechanistically stabilizing nNOS-multiprotein complex at the myofiber sarcolemma.


Subject(s)
Membrane Glycoproteins/metabolism , Muscular Disorders, Atrophic/metabolism , Muscular Disorders, Atrophic/pathology , Nitric Oxide Synthase Type I/metabolism , Sarcolemma/enzymology , Animals , Enzyme Stability , Female , Muscular Disorders, Atrophic/enzymology , Rats , Rats, Wistar , Sarcolemma/metabolism
6.
Mol Med ; 18: 1261-8, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22952056

ABSTRACT

Spinal and bulbar muscular atrophy is an X-linked motor neuron disease caused by polyglutamine expansion in the androgen receptor. Patients develop slowly progressive proximal muscle weakness, muscle atrophy and fasciculations. Affected individuals often show gynecomastia, testicular atrophy and reduced fertility as a result of mild androgen insensitivity. No effective disease-modifying therapy is currently available for this disease. Our recent studies have demonstrated that insulinlike growth factor (IGF)-1 reduces the mutant androgen receptor toxicity through activation of Akt in vitro, and spinal and bulbar muscular atrophy transgenic mice that also overexpress a noncirculating muscle isoform of IGF-1 have a less severe phenotype. Here we sought to establish the efficacy of daily intraperitoneal injections of mecasermin rinfabate, recombinant human IGF-1 and IGF-1 binding protein 3, in a transgenic mouse model expressing the mutant androgen receptor with an expanded 97 glutamine tract. The study was done in a controlled, randomized, blinded fashion, and, to reflect the clinical settings, the injections were started after the onset of disease manifestations. The treatment resulted in increased Akt phosphorylation and reduced mutant androgen receptor aggregation in muscle. In comparison to vehicle-treated controls, IGF-1-treated transgenic mice showed improved motor performance, attenuated weight loss and increased survival. Our results suggest that peripheral tissue can be targeted to improve the spinal and bulbar muscular atrophy phenotype and indicate that IGF-1 warrants further investigation in clinical trials as a potential treatment for this disease.


Subject(s)
Insulin-Like Growth Factor I/therapeutic use , Muscular Disorders, Atrophic/drug therapy , Muscular Disorders, Atrophic/pathology , Animals , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/pathology , Muscles/drug effects , Muscles/metabolism , Muscles/pathology , Muscles/physiopathology , Muscular Disorders, Atrophic/enzymology , Muscular Disorders, Atrophic/physiopathology , Mutant Proteins/metabolism , Phosphorylation/drug effects , Protein Structure, Quaternary , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/metabolism , Weight Loss/drug effects
7.
Muscle Nerve ; 43(5): 708-16, 2011 May.
Article in English | MEDLINE | ID: mdl-21462205

ABSTRACT

INTRODUCTION: Our goal was to determine whether in vivo administration of the proteasome inhibitor MG132 can prevent muscle atrophy caused by hindlimb unloading (HU). METHODS: Twenty-seven NMRI mice were assigned to a weight-bearing control, a 6-day HU, or a HU+MG132 (1 mg/kg/48 h) treatment group. RESULTS: Gastrocnemius wasting was significantly less in HU+MG132 mice (-6.7 ± 2.0%) compared with HU animals (-12.6 ± 1.1%, P = 0.011). HU was also associated with an increased expression of MuRF-1 (P = 0.006), MAFbx (P = 0.001), and USP28 (P = 0.027) mRNA, whereas Nedd4, E3α, USP19, and UBP45 mRNA did not change significantly. Increases in MuRF-1, MAFbx, and USP28 mRNA were largely repressed after MG132 administration. ß5 proteasome activity tended to increase in HU (+16.7 ± 6.1%, P = 0.086). Neither ß1 and ß2 proteasome activities nor ubiquitin-conjugated proteins were changed by HU. CONCLUSIONS: Our results indicate that in vivo administration of MG132 partially prevents muscle atrophy associated with disuse and highlight an unexpected regulation of MG132 proteasome inhibitor on ubiquitin-ligases.


Subject(s)
Cysteine Proteinase Inhibitors/therapeutic use , Leupeptins/therapeutic use , Muscular Disorders, Atrophic/drug therapy , Muscular Disorders, Atrophic/enzymology , Proteasome Inhibitors , Animals , Cysteine Proteinase Inhibitors/pharmacology , Hindlimb Suspension/methods , Leupeptins/pharmacology , Male , Mice , Muscular Atrophy/drug therapy , Muscular Atrophy/enzymology , Proteasome Endopeptidase Complex/metabolism , Random Allocation
8.
Histochem Cell Biol ; 132(4): 383-94, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19644701

ABSTRACT

We monitored changes in SERCA isoform specific expression and S-nitrosylation in myofibers of lower limb soleus (SOL) and vastus lateralis (VL) muscle biopsies before and after 60 days of voluntary long term bed rest (BR) without (BR-CTRL group, n = 8) and with exercise countermeasure (BR-EX group, n = 8). Before BR, a typical myofiber type-specific distribution of fast and slow SERCA1/2a isoforms was seen. After BR, a subpopulation (approx. 15%) of slow myofibers in BR-CTRL additionally expressed the fast SERCA1a isoform which was not seen in BR-EX. After BR, SERCA1a S-nitrosylation patterns analyzed by the biotin-switch assay decreased in disused SOL only but increased in both muscles following exercise. Differential SERCA1a S-nitrosylation and SERCA1a/2a co-expression in subsets of slow myofibers should be considered as signs of an altered cytosolic Ca(2+) homeostasis following chronic muscle disuse. Exercise preserved myofiber type-specific SERCA1a expression and S-nitrosylation in VL and SOL in a different way, suggesting muscle-specific responses to the countermeasure protocol applied during bed rest.


Subject(s)
Muscle Fibers, Slow-Twitch/enzymology , Muscular Disorders, Atrophic/enzymology , Myofibrils/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Bed Rest/adverse effects , Exercise/physiology , Female , Humans , Lower Extremity , Muscular Disorders, Atrophic/pathology , Muscular Disorders, Atrophic/prevention & control , Protein Isoforms/metabolism , Quadriceps Muscle/enzymology
9.
J Biol Chem ; 283(12): 7648-56, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18201976

ABSTRACT

The accumulation of intracellular protein deposits as inclusion bodies is the common pathological hallmark of most age-related neurodegenerative disorders including polyglutamine diseases. Appearance of aggregates of the misfolded mutant disease proteins suggest that cells are unable to efficiently degrade them, and failure of clearance leads to the severe disturbances of the cellular quality control system. Recently, the quality control ubiquitin ligase CHIP has been shown to suppress the polyglutamine protein aggregation and toxicity. Here we have identified another ubiquitin ligase, called E6-AP, which is able to promote the proteasomal degradation of misfolded polyglutamine proteins and suppress the polyglutamine protein aggregation and polyglutamine protein-induced cell death. E6-AP interacts with the soluble misfolded polyglutamine protein and associates with their aggregates in both cellular and transgenic mouse models. Partial knockdown of E6-AP enhances the rate of aggregate formation and cell death mediated by the polyglutamine protein. Finally, we have demonstrated the up-regulation of E6-AP in the expanded polyglutamine protein-expressing cells as well as cells exposed to proteasomal stress. These findings suggest that E6-AP is a critical mediator of the neuronal response to misfolded polyglutamine proteins and represents a potential therapeutic target in the polyglutamine diseases.


Subject(s)
Disease Models, Animal , Peptides/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , COS Cells , Cell Death/genetics , Chlorocebus aethiops , Humans , Huntington Disease/enzymology , Huntington Disease/genetics , Mice , Mice, Transgenic , Muscular Disorders, Atrophic/enzymology , Muscular Disorders, Atrophic/genetics , Peptides/genetics , Proteasome Endopeptidase Complex/genetics , Protein Folding , Ubiquitin-Protein Ligases/genetics
10.
J Clin Invest ; 117(12): 3940-51, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17992259

ABSTRACT

Statins inhibit HMG-CoA reductase, a key enzyme in cholesterol synthesis, and are widely used to treat hypercholesterolemia. These drugs can lead to a number of side effects in muscle, including muscle fiber breakdown; however, the mechanisms of muscle injury by statins are poorly understood. We report that lovastatin induced the expression of atrogin-1, a key gene involved in skeletal muscle atrophy, in humans with statin myopathy, in zebrafish embryos, and in vitro in murine skeletal muscle cells. In cultured mouse myotubes, atrogin-1 induction following lovastatin treatment was accompanied by distinct morphological changes, largely absent in atrogin-1 null cells. In zebrafish embryos, lovastatin promoted muscle fiber damage, an effect that was closely mimicked by knockdown of zebrafish HMG-CoA reductase. Moreover, atrogin-1 knockdown in zebrafish embryos prevented lovastatin-induced muscle injury. Finally, overexpression of PGC-1alpha, a transcriptional coactivator that induces mitochondrial biogenesis and protects against the development of muscle atrophy, dramatically prevented lovastatin-induced muscle damage and abrogated atrogin-1 induction both in fish and in cultured mouse myotubes. Collectively, our human, animal, and in vitro findings shed light on the molecular mechanism of statin-induced myopathy and suggest that atrogin-1 may be a critical mediator of the muscle damage induced by statins.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Lovastatin/adverse effects , Muscle Proteins/metabolism , Muscular Disorders, Atrophic/enzymology , SKP Cullin F-Box Protein Ligases/metabolism , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cholesterol/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Mice , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Disorders, Atrophic/chemically induced , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , SKP Cullin F-Box Protein Ligases/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
11.
Am J Physiol Regul Integr Comp Physiol ; 291(6): R1730-40, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16873557

ABSTRACT

Skeletal muscle atrophy is associated with an increase in apoptosis, and we showed previously that endonuclease G (EndoG) is localized to nuclei following unloading. The goal of this study was to determine whether the onset of apoptosis in response to disuse was consistent with the hypothesis that EndoG is involved in myofiber nuclear loss. Atrophy was induced by hindlimb suspension for 12 h or 1, 2, 4 and 7 days in 6-mo-old rats. Soleus myofiber cross-sectional area decreased significantly by 2 days, whereas muscle mass and muscle-to-body mass ratio decreased by 4 and 7 days, respectively. By contrast, a significant increase in apoptosis, evidenced by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive nuclei, occurred as early as 12 h after suspension, preceding the elevation in muscle atrophy F-box gene expression. The early increase in apoptosis appeared to be specific to myofiber nuclei, whereas TUNEL-positive interstitial cells did not become significantly elevated until 2 days after suspension. Furthermore, TUNEL-positive myofiber nuclei colocalized with EndoG as early as 12 h after suspension, and no such localization was observed in interstitial cells. Although no significant change in total activated caspase-3, -7, or -12 protein abundance was apparent, activated caspase-3 was expressed in interstitial cells undergoing apoptosis, some of which were endothelial cells. These data indicate that apoptosis is an early, and therefore possibly causative, event in the process of muscle atrophy, and that EndoG nuclear translocation is specific for myofiber nuclear apoptosis, whereas interstitial cells may undergo apoptosis via a more classical, caspase-dependent pathway.


Subject(s)
Cell Nucleus/metabolism , Endodeoxyribonucleases/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Disorders, Atrophic/enzymology , Muscular Disorders, Atrophic/pathology , Active Transport, Cell Nucleus , Animals , Apoptosis , Cell Nucleus/pathology , Hindlimb Suspension/adverse effects , Male , Muscular Disorders, Atrophic/etiology , Rats , Rats, Sprague-Dawley
12.
Histol Histopathol ; 20(1): 99-106, 2005 01.
Article in English | MEDLINE | ID: mdl-15578428

ABSTRACT

Muscle atrophy commonly occurs as a consequence of prolonged muscle inactivity, as observed after cast immobilization, bed rest or space flights. The molecular mechanisms responsible for muscle atrophy are still unknown, but a role has been proposed for altered permeability of the sarcolemma and of the surrounding connective tissue. Matrix metallo-proteinases (MMPs) are a family of enzymes with proteolytic activity toward a number of extracellular matrix (ECM) components; they are inhibited by tissue inhibitors of MMPs (TIMPs). In a rat tail-suspension experimental model, we show that after fourteen days of non-weight bearing there is increased expression of MMP-2 in the atrophic soleus and gastrocnemius and decreased expression of TIMP-2. In the same experimental model the expression of Collagen I and Collagen IV, two main ECM components present in the muscles, was reduced and unevenly distributed in unloaded animals. The difference was more evident in the soleus than in the gastrocnemius muscle. This suggests that muscle disuse induces a proteolytic imbalance, which could be responsible for the breakdown of basal lamina structures such as Collagen I and Collagen IV, and that this leads to an altered permeability with consequent atrophy. In conclusion, an MMP-2/TIMP-2 imbalance could have a role in the mechanism underlying muscle disuse atrophy; more studies are needed to expand our molecular knowledge on this issue and to explore the possibility of targeting the proteolytic imbalance with MMP inhibitors.


Subject(s)
Matrix Metalloproteinases/metabolism , Muscle, Skeletal/enzymology , Muscular Disorders, Atrophic/enzymology , Animals , Fluorescent Antibody Technique , Male , Matrix Metalloproteinase 2/metabolism , Rats , Rats, Wistar , Time Factors , Tissue Inhibitor of Metalloproteinase-2/metabolism
13.
Am J Clin Pathol ; 116(3): 326-30, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11554158

ABSTRACT

We retrospectively reviewed 118 muscle biopsy specimens from 113 patients with clinical and/or biochemical evidence of mitochondrial cytopathy. Light microscopic evaluation revealed histologic abnormalities in 65 specimens. The most common histologic findings included angular atrophic esterase-positive muscle fibers, type II muscle atrophy, regenerating muscle fibers, and scattered cytochrome-oxidase deficient fibers. Ragged red fibers were noted in 3 specimens on a Gomori trichrome stain. Electron microscopic evaluation was performed in 113 muscle specimens, and in 34, no abnormalities were identified. Increased numbers of mitochondria, particularly in the subsarcolemmal region, were identified in 54 specimens. Increased mitochondrial size was seen in 8 specimens and paracrystalline mitochondrial inclusions in 3. Other ultrastructural findings included focally increased glycogen deposition, focal Z-band streaming, and focally increased lipid accumulation. For 39 cases, concomitant skin biopsy specimens were available; abnormalities were identified by electron microscopy in 12. The majority of biopsy specimens demonstrated some light or electron microscopic abnormality. Specific histologic findings suggestive of mitochondrial abnormalities (partial cytochrome oxidase deficiency, ragged red fibers) were noted in a minority of cases. Ultrastructural evidence of mitochondrial abnormalities was noted in the majority of cases.


Subject(s)
Mitochondria/ultrastructure , Mitochondrial Myopathies/pathology , Muscle, Skeletal/pathology , Adolescent , Adult , Aged , Biopsy , Child , Child, Preschool , Electron Transport Complex IV/metabolism , Esterases/metabolism , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mitochondrial Myopathies/enzymology , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/enzymology , Muscular Atrophy/enzymology , Muscular Atrophy/pathology , Muscular Disorders, Atrophic/enzymology , Muscular Disorders, Atrophic/pathology , Retrospective Studies , Skin/enzymology , Skin/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...