Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
Add more filters











Publication year range
1.
Neuropathol Appl Neurobiol ; 39(7): 750-61, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23452232

ABSTRACT

AIMS: Sporadic inclusion-body myositis (s-IBM) is an age-associated degenerative muscle disease. Characteristic features are muscle-fibre vacuolization and intramuscle-fibre accumulations of multiprotein aggregates, which may result from the demonstrated impairments of the 26S proteasome and autophagy. Chaperone-mediated autophagy (CMA) is a selective form of lysosomal degradation targeting proteins carrying the KFERQ motif. Lysosome-associated membrane protein type 2A (LAMP2A) and the heat-shock cognate protein 70 (Hsc70) constitute specific CMA components. Neither CMA components nor CMA activity has been studied in normal or disease human muscle, to our knowledge. METHODS: We studied CMA components by immunocytochemistry, immunoblots, real-time PCR and immunoprecipitation in: (a) 16 s-IBM, nine aged-matched normal and nine disease control muscle biopsies; and (b) cultured human muscle fibres (CHMFs) with experimentally inhibited activities of either the 26S proteasome or autophagy. RESULTS: Compared with age-matched controls, in s-IBM muscle, LAMP2A and Hsc70 were on a given transverse section accumulated as aggregates in approximately 5% of muscle fibres, where they (a) colocalized with each other and α-synuclein (α-syn), a CMA-targeted protein; and (b) were bound to each other and to α-syn by immunoprecipitation. By immunoblots, LAMP2A was increased sevenfold P < 0.001 and Hsc70 2.6-fold P < 0.05. LAMP2A mRNA was increased 4.4-fold P < 0.001 and Hsc70 mRNA 1.9-fold P < 0.05. In CHMFs inhibition of either the 26S proteasome or autophagy induced CMA, evidenced by a significant increase of both LAMP2A and Hsc70. CONCLUSIONS: Our study demonstrates, for the first time, up-regulation of CMA components in s-IBM muscle, and it provides further evidence that altered protein degradation is likely an important pathogenic aspect in s-IBM.


Subject(s)
Autophagy/physiology , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Muscle Fibers, Skeletal/metabolism , Myositis, Inclusion Body/metabolism , Aged , Aged, 80 and over , Female , Humans , Lysosomes/metabolism , Male , Middle Aged , Muscle Fibers, Skeletal/pathology , Myositis, Inclusion Body/pathology , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/metabolism , Up-Regulation/physiology , alpha-Synuclein/metabolism
3.
Folia Morphol (Warsz) ; 67(1): 6-12, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18335407

ABSTRACT

Preferential atrophy of Type-II muscle fibres occurs in several clinical situations, including cachexia, muscle disuse, chronic glucocorticoid treatment, remote neoplasia, and sometimes as an aspect of recent-denervation. For the patient, the Type-II atrophy itself might be unfavourable (as a glucocorticoid side-effect) or favourable (survivalistic via the muscle-alanine liver-gluconeogenesis pathway in starvation). The cellular mechanisms underlying Type-II fibre atrophy are unclear. Myostatin (Mstn) is physiologically a negative regulator of muscle mass and strength. In this study we evaluated a possible role of Mstn in Type-II fibre atrophy in human muscle. Mstn and Mstn precursor protein (MstnPP) were studied in 10-muscle biopsies containing Type-II fibre atrophy and in 17 disease and normal control muscle biopsies. When comparison was made with normal control fibres, we found the following: 1) by immunocytochemistry, diffusely increased Mstn/MstnPP in the atrophic Type-II muscle fibres; 2) by immunoblots, Mstn/MstnPP increased individually; 3) by RT-PCR, no increase in MstnPP mRNA. In conclusion, our results a) suggest that Mstn/ /MstnPP might play a role in the pathogenic cascade of Type-II muscle fibre atrophy; b) broaden our previously-described associations of Mstn in human muscle pathology, and c) could possibly lead to clinical prevention when Type-II muscle fibre atrophy is unfavourable, for instance in glucocorticoid therapy.


Subject(s)
Muscle Fibers, Fast-Twitch/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Protein Precursors/metabolism , Transforming Growth Factor beta/metabolism , Adenosine Triphosphatases/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers/analysis , Biomarkers/metabolism , Biopsy , Glucocorticoids/adverse effects , Humans , Immunohistochemistry , Middle Aged , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Myostatin , Protein Precursors/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta/genetics , Up-Regulation/physiology
5.
Acta Myol ; 25(1): 13-22, 2006 Jun.
Article in English | MEDLINE | ID: mdl-17039976

ABSTRACT

UNLABELLED: Parkin, an E3-ubiquitin ligase in the ubiquitin-proteasome system, facilitates degradation of alpha-synuclein and other proteins. Since ubiquitinated multiprotein-aggregates containing amyloid-beta (Abeta), alpha-synuclein, and other proteins, are characteristic of sporadic inclusion-body myositis (s-IBM) muscle fibers, we asked whether parkin might have a role in s-IBM pathogenesis. We studied the association of parkin with alpha-synuclein and Abeta-precursor protein (AbetaPP) in s-IBM muscle biopsies and in our IBM model based on overexpression of AbetaPP into cultured human muscle fibers. We report the following in s-IBM muscle fibers: a) parkin was increased 2.7 fold and accumulated in aggregates also containing Abeta and alpha-synuclein; b) alpha-synuclein was increased 6.3 fold; c) parkin physically associated with alpha-synuclein and AbetaPP; d) alpha-synuclein and AbetaPP were ubiquitinated. In the IBM model: a) parkin was increased 2.7 fold, b) it associated with alpha-synuclein and AbetaPP. CONCLUSION: 1. This is the first demonstration that in a human muscle disease alpha-synuclein associates with parkin, and might be ubiquitinated by it. 2. The small increase of parkin relative to the much larger increase of alpha-synuclein might be insufficient to secure complete ubiquitination and consequent degradation of alpha-syn. 3. AbetaPP might be a novel substrate of parkin.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Muscle Fibers, Skeletal/metabolism , Myositis, Inclusion Body/metabolism , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/physiology , Case-Control Studies , Humans , Myositis, Inclusion Body/etiology , Myositis, Inclusion Body/pathology , Tissue Culture Techniques
6.
Acta Myol ; 25(3): 116-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17626519

ABSTRACT

Nogo (RTN4) belongs to the reticulon (RTN) family of integral membrane proteins. RTN4A (Nogo-A), RTN4B (Nogo-B) and RTN4C (Nogo-C) are isoforms of RTN4. In the gastrocnemius muscle of transgenic mice bearing an SOD1 mutation ("ALS model"), increased Nogo-A mRNA and protein was reported, and similar changes were reported in muscle biopsies of patients with amyotrophic lateral sclerosis (ALS) but not with peripheral neuropathy or primary muscle diseases, leading to the proposal that Nogo-A in skeletal muscle is a new specific molecular marker of ALS. Here we report, based on studies of muscle biopsies from patients with ALS, peripheral neuropathies, polymyositis, dermatomyositis and morphologically nonspecific myopathies that, in addition of strong Nogo-A immunoreactivity within apparently-denervated small angular fibers in ALS and peripheral neuropathies, Nogo-A was strongly immunoreactive within desmin-positive regenerating muscle fibers in various myopathies, and its expression on immunoblots was increased in all those neuromuscular diseases. In conclusion, we have found that the presence of Nogo-A in diseased human muscle biopsies is not limited to ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Muscle, Skeletal/metabolism , Myelin Proteins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Biopsy , Dermatomyositis/metabolism , Dermatomyositis/pathology , Gene Expression Regulation , Humans , Muscle, Skeletal/pathology , Myelin Proteins/genetics , Nogo Proteins , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Polymyositis/metabolism , Polymyositis/pathology
7.
Acta Myol ; 24(1): 17-24, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16315367

ABSTRACT

Sporadic inclusion-body myositis (s-IBM), the most common muscle disease of older persons, is of unknown cause and there is no successful treatment. We summarize our most recent findings in s-IBM muscle fibers, which demonstrate abnormalities of the ubiquitin-proteasome system, and abnormal accumulation, misfolding and aggregation of proteins. We propose that these changes, possibly provoked by the aging intra-muscle fiber cellular milieu, and aggravated by the oxidative stress, play a key pathogenic role in s-IBM. This evidence strongly suggests that mechanisms other than the immune/inflammatory response play the important role in s-IBM muscle fiber degeneration.


Subject(s)
Myositis, Inclusion Body/etiology , Proteasome Endopeptidase Complex/physiology , Protein Folding , Ubiquitin/physiology , Adenosine Triphosphatases , Cell Cycle Proteins/physiology , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/physiology , Humans , Muscle Fibers, Skeletal/metabolism , Mutation/physiology , Valosin Containing Protein
8.
Neurology ; 63(6): 1114-7, 2004 Sep 28.
Article in English | MEDLINE | ID: mdl-15452314

ABSTRACT

Mutant ubiquitin (UBB+1), a product of "molecular misreading," is toxic to cells because its ubiquitinated form inhibits the proteasome, contributing to accumulation of misfolded proteins and their ensuing toxicity. The authors demonstrate in 10 sporadic inclusion body myositis (s-IBM) muscle biopsies that UBB+1 is accumulated in aggregates containing amyloid-beta and phosphorylated-tau. In s-IBM, UBB+1 may be pathogenic by inhibiting proteasome, thereby promoting accumulation of cytotoxic misfolded amyloid-beta and phosphorylated-tau.


Subject(s)
Muscle Fibers, Skeletal/metabolism , Myositis, Inclusion Body/genetics , Ubiquitin/genetics , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , Biopsy , Humans , Microscopy, Fluorescence , Microscopy, Immunoelectron , Molecular Sequence Data , Muscular Diseases/metabolism , Muscular Diseases/pathology , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Phosphorylation , Proteasome Inhibitors , Protein Folding , Protein Processing, Post-Translational , Ubiquitin/metabolism , tau Proteins/metabolism
9.
Neurology ; 58(3): 438-45, 2002 Feb 12.
Article in English | MEDLINE | ID: mdl-11839845

ABSTRACT

BACKGROUND: An important aspect of inclusion-body myositis (IBM) vacuolated muscle fibers (VMF) is abnormal accumulation of amyloid-beta precursor protein (AbetaPP) epitopes and its product, amyloid-beta (Abeta), and of phosphorylated tau (p-tau) in the form of paired helical filaments. Lipoprotein receptors and cholesterol are known to play an important role in AbetaPP processing, Abeta production, and tau phosphorylation. METHODS: In 10 IBM and 22 control muscle biopsies the authors immunolocalized low-density lipoprotein receptor (LDLR), very low-density lipoprotein receptor (VLDLR), and low-density lipoprotein receptor-related protein (LRP), and colocalized them with Abeta, p-tau, APOE, and free cholesterol. RESULTS: In each biopsy, virtually all IBM VMF had strong LDLR-immunoreactive inclusions, which colocalized with Abeta, APOE, p-tau, and free cholesterol. VLDLR was increased mainly diffusely, but in approximately 50% of the VMF it was also accumulated in the form of inclusions colocalizing with Abeta, APOE, and free cholesterol, but not with p-tau. LRP inclusions were present in a few VMF. In all myopathies, a subset of regenerating and necrotizing muscle fibers had prominent diffuse accumulation of both LDLR and free cholesterol. At normal neuromuscular junctions (NMJ) postsynaptically, LDLR and VLDLR, but not LRP, were immunoreactive. CONCLUSIONS: 1) Abnormal accumulation of LDLR, VLDLR, LRP, and cholesterol within IBM vacuolated muscle fibers suggests novel roles for them in the IBM pathogenesis. 2) Expression of LDLR and VLDLR at normal NMJ suggests physiologic roles for them in transsynaptic signaling pathways, increased internalization of lipoproteins there, or both. 3) Increased LDLR and free cholesterol in some regenerating and necrotizing muscle fibers suggest a role for them in human muscle fiber growth and repair and necrotic death.


Subject(s)
Cholesterol/analysis , Muscle, Skeletal/chemistry , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Receptors, Lipoprotein/analysis , Amyloid beta-Peptides/analysis , Apolipoproteins E/analysis , Biopsy , Humans , Low Density Lipoprotein Receptor-Related Protein-1/analysis , Microscopy, Immunoelectron , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/pathology , Phosphorylation , Receptors, LDL/analysis , tau Proteins/analysis , tau Proteins/metabolism
10.
Lancet ; 358(9297): 1962-4, 2001 Dec 08.
Article in English | MEDLINE | ID: mdl-11747923

ABSTRACT

Sporadic inclusion-body myositis (IBM) is the most common, progressive muscle disease of older individuals. We investigated the presence of BACE1 and BACE2-two beta secretases that cleave amyloid-beta-precursor protein-in muscle-biopsy samples from patients with IBM and from controls. On immunofluorescence, BACE1 and BACE2 co-localised with amyloid beta in IBM vacuolated muscle fibres, but were not found in controls. Immunoblotting showed increased BACE2 but not BACE1 in patients with IBM compared with controls. Our study suggests that both of these proteases might participate in processing of amyloid-beta-precursor protein in IBM muscle fibres.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Myositis, Inclusion Body/enzymology , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Biopsy , Case-Control Studies , Endopeptidases , Fluorescent Antibody Technique , Humans , Immunoblotting , Muscle Fibers, Skeletal/enzymology
11.
Neuroreport ; 12(9): 1809-14, 2001 Jul 03.
Article in English | MEDLINE | ID: mdl-11435903

ABSTRACT

Sporadic inclusion-body myositis (IBM) is a progressive degenerative muscle disease of older persons. Abnormalities of gene-expression and RNA metabolism have recently been proposed to contribute to the IBM pathogenic cascade. We now demonstrate, using well characterized, epitope-specific antibodies, that the largest subunit of RNA polymerase II is abnormally accumulated in the cytoplasm of IBM muscle fibers, where it is co-localized with phosphorylated tau on IBM paired helical filaments. Since RNA polymerase II is a crucial nuclear factor involved in both transcription and mRNA processing, our results support the hypothesis that abnormality of either or both of those processes might be caused, in part, by pathological trafficking of RNA polymerase II, and that abnormal trafficking might be an important factor in the IBM pathogenic cascade.


Subject(s)
Cytoplasm/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/enzymology , Myositis, Inclusion Body/enzymology , RNA Polymerase II/metabolism , Cytoplasm/pathology , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic/genetics , Humans , Microscopy, Electron , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myositis, Inclusion Body/pathology , Myositis, Inclusion Body/physiopathology , Protein Transport/genetics , RNA Polymerase II/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/genetics
12.
Proc Natl Acad Sci U S A ; 98(7): 4022-7, 2001 Mar 27.
Article in English | MEDLINE | ID: mdl-11274426

ABSTRACT

The recently discovered aging-dependent large accumulation of point mutations in the human fibroblast mtDNA control region raised the question of their occurrence in postmitotic tissues. In the present work, analysis of biopsied or autopsied human skeletal muscle revealed the absence or only minimal presence of those mutations. By contrast, surprisingly, most of 26 individuals 53 to 92 years old, without a known history of neuromuscular disease, exhibited at mtDNA replication control sites in muscle an accumulation of two new point mutations, i.e., A189G and T408A, which were absent or marginally present in 19 individuals younger than 34 years. These two mutations were not found in fibroblasts from 22 subjects 64 to 101 years of age (T408A), or were present only in three subjects in very low amounts (A189G). Furthermore, in several older individuals exhibiting an accumulation in muscle of one or both of these mutations, they were nearly absent in other tissues, whereas the most frequent fibroblast-specific mutation (T414G) was present in skin, but not in muscle. Among eight additional individuals exhibiting partial denervation of their biopsied muscle, four subjects >80 years old had accumulated the two muscle-specific point mutations, which were, conversely, present at only very low levels in four subjects < or =40 years old. The striking tissue specificity of the muscle mtDNA mutations detected here and their mapping at critical sites for mtDNA replication strongly point to the involvement of a specific mutagenic machinery and to the functional relevance of these mutations.


Subject(s)
Aging/genetics , DNA, Mitochondrial/genetics , Muscles/physiology , Point Mutation , Adult , Age Distribution , Aged , Aged, 80 and over , DNA Replication/genetics , Female , Fibroblasts/physiology , Humans , Male , Middle Aged
13.
J Neuropathol Exp Neurol ; 60(1): 1-14, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11202170

ABSTRACT

We review the newest advances related to seeking the pathogenic mechanism(s) of sporadic inclusion-body myositis (s-IBM) and present the pathologic diagnostic criteria of s-IBM. We discuss the possible pathogenic role of several themes, such as 1) increased amyloid-beta precursor protein (AbetaPP) and of its fragment Abeta; 2) phosphorylation of tau protein; 3) oxidative stress; 4) abnormal a) signal-transduction, b) transcription, and c) RNA accumulation; 5) "junctionalization" and myogenous" denervation; and 6) lymphocytic inflammation. Evidence is provided supporting our hypothesis that overexpression of AbetaPP within the aging muscle fibers is an early upstream event causing the subsequent pathogenic cascade. The remarkable pathologic similarities between s-IBM muscle and Alzheimer disease (AD) brain are discussed, and the possible cause and significance are addressed.


Subject(s)
Aging/physiology , Alzheimer Disease/pathology , Myositis, Inclusion Body/etiology , Myositis, Inclusion Body/pathology , Brain/pathology , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myositis, Inclusion Body/metabolism
14.
Neurosci Lett ; 293(1): 33-6, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11065131

ABSTRACT

To investigate the possible role of cyclin-dependent kinase 5 (cdk5) in the formation of paired helical filaments (PHFs) in muscle of patients with inclusion-body myositis (IBM), we immunolocalized cdk5, by light- and electron- microscopy, in muscle biopsies of six IBM patients. Approximately 80-90% of IBM vacuolated muscle fibers, and 10-15% of nonvacuolated fibers, contained well defined cdk5-immunoreactive inclusions that colocalized with phosphorylated tau in 70-80% of those fibers. Immunoelectronmicroscopy revealed the association of cdk5 with tau-immunoreactive PHFs. In all biopsies that contained them, regenerating muscle fibers had diffuse, moderate to strong cdk5 immunoreactivity. At all neuromuscular junctions, there was strong cdk5 immunoreactivity postsynaptically. Our study suggests that cdk5: (1) plays a role in IBM pathogenesis, possibly mediating phosphorylation of PHF-related tau; (2) is involved in muscle regeneration; and (3) has a novel function at normal neuromuscular junctions.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Muscle, Skeletal/metabolism , Myositis, Inclusion Body/metabolism , tau Proteins/metabolism , Cyclin-Dependent Kinase 5 , Humans , Microscopy, Immunoelectron , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiopathology , Muscle, Skeletal/ultrastructure , Myositis, Inclusion Body/pathology , Neuromuscular Junction/metabolism , Phosphorylation , Regeneration/physiology , Tissue Distribution , Vacuoles/metabolism
15.
Neuromuscul Disord ; 10(8): 604-11, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11053689

ABSTRACT

Mitochondrial DNA variants have been shown to be associated with many diseases. Mutations at mitochondrial DNA nucleotide positions 3192, 3196, 3397 and 4336 have been described in association with late-onset Alzheimer's disease. The pathological similarities between inclusion body myositis and Alzheimer's disease prompted an analysis of the relationship between the reported mutations and sporadic inclusion body myositis. The 4336G variant was not significantly increased in patients with inclusion body myositis or Alzheimer's disease when compared to controls. None of the patients with inclusion body myositis carried mutations at nucleotide positions 3192, 3196 and 3397. A transition at nucleotide position 4580 was detected in some patients with inclusion body myositis and Alzheimer's disease but was not significantly higher in frequency when compared to controls. Phylogenetic analysis showed that the 4336G and 4580A variants clustered together in their respective group. A group of patients with inclusion body myositis also clustered together on a separate branch of the phylogenetic tree. Closer investigation of this group revealed a common polymorphism at nucleotide position 16311. The frequency of the 16311C variant was higher in inclusion body myositis than in Alzheimer's disease and controls, although when only caucasian patients were considered the increased frequency was not statistically significant. Further studies will be required to determine whether this variant plays a role in the pathogenesis of inclusion body myositis.


Subject(s)
DNA Mutational Analysis/statistics & numerical data , DNA, Mitochondrial/genetics , Mutation/genetics , Myositis, Inclusion Body/genetics , Adult , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Evolution, Molecular , Humans , Middle Aged , Multigene Family/genetics , Phylogeny
16.
J Neuropathol Exp Neurol ; 59(7): 592-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10901230

ABSTRACT

Alpha-synuclein (alpha-syn) is an important component of neuronal and glial inclusions in brains of patients with several neurodegenerative disorders. Sporadic inclusion-body myositis (s-IBM) is the most common progressive muscle disease of older patients. Its muscle phenotype shows several similarities with Alzheimer disease brain. A distinct feature of s-IBM pathology is specific vacuolar degeneration of muscle fibers characterized by intracellular amyloid inclusions formed by both amyloid-beta (Abeta) and paired-helical filaments composed of phosphorylated tau. We immunostained alpha-syn in muscle biopsies of s-IBM, disease-control, and normal patients. Approximately 60% of Abeta-positive vacuolated muscle fibers (VMF) contained well-defined inclusions immunoreactive with antibodies against alpha-syn. In those fibers. alpha-syn co-localized with Abeta, both by light microscopy, and ultrastructurally. Paired-helical filaments did not contain alpha-syn immunoreactivity. In all muscle biopsies, alpha-syn was strongly immunoreactive at the postsynaptic region of the neuromuscular junctions. alpha-syn immunoreactivity also occurred diffusely in regenerating and necrotic muscle fibers. In cultured human muscle fibers, alpha-syn and its mRNA were expressed by immunocytochemistry, immunoblots, and Northern blots. Our study provides the first demonstration that alpha-syn participates in normal and pathologic processes of human muscle. Therefore. its function is not exclusive to the brain and neurodegenerative diseases.


Subject(s)
Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Myositis, Inclusion Body/pathology , Nerve Tissue Proteins/analysis , Neuromuscular Junction/pathology , Cells, Cultured , Fluorescent Antibody Technique , Humans , Inclusion Bodies/chemistry , Inclusion Bodies/pathology , Microscopy, Immunoelectron , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/physiology , Necrosis , Neuromuscular Junction/ultrastructure , Regeneration , Synucleins , alpha-Synuclein
17.
Am J Pathol ; 156(6): 1835-40, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10854206

ABSTRACT

The possible role of extracellular signal-regulated kinase (ERK) in the pathogenesis of inclusion-body myositis (IBM) was investigated by immunostaining the active phosphorylated form of ERK in muscle biopsies of six IBM and 14 control patients. Between 80% and 90% of IBM vacuolated muscle fibers contained well-defined ERK-immunoreactive inclusions, which were co-localized by light microscopy, with phosphorylated tau in 70 to 80% of those fibers. Immunoelectronmicroscopy colocalized ERK to small amorphous tufts adjacent to the muscle fiber paired-helical filaments. Strong ERK immunoreactivity was also present at the postsynaptic domain of all human neuromuscular junctions. Our study suggests 1) that ERK, a signal transducer, might play a role in IBM pathogenesis, including participation in the pathological phosphorylation of IBM tau; and 2) that signal transduction abnormalities may be a component of the IBM pathogenic cascade. Our novel immunolocalization of ERK at the postsynaptic domain of human neuromuscular junctions supports a role in transcription of junctional-protein genes. The ERK localized in nonjunctional regions of IBM fibers may underlie the known pathological up-regulation of junctional proteins there.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Muscles/metabolism , Myositis, Inclusion Body/metabolism , Neuropil Threads/metabolism , tau Proteins/metabolism , Humans , Immunohistochemistry , Microscopy, Immunoelectron , Myositis, Inclusion Body/pathology , Phosphorylation , Reference Values
18.
Neurosci Lett ; 287(1): 1-4, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10841976

ABSTRACT

To determine whether redox factor-1 (Ref-1) participates in the pathogenesis of inclusion-body myositis (IBM), we immunolocalized Ref-1 in muscle biopsies of IBM patients by light- and electron-microscopy. Approximately 70-80% of the IBM vacuolated muscle fibers had focal inclusions strongly immunoreactive for Ref-1. By immunoelectronmicroscopy, Ref-1 was localized to paired-helical filaments, 6-10 nm amyloid-like fibrils and amorphous material. Virtually all regenerating and necrotic muscle fibers in various muscle biopsies had diffusely strong Ref-1 immunoreactivity. At all neuromuscular junctions, postsynaptically there was strong Ref-1 immunoreactivity. Our study suggests that Ref-1 plays a role in IBM pathogenesis, and in other pathologic and normal processes of human muscle.


Subject(s)
Carbon-Oxygen Lyases/analysis , DNA-(Apurinic or Apyrimidinic Site) Lyase , Muscle, Skeletal/pathology , Myositis, Inclusion Body/pathology , Biopsy , Humans , Microscopy, Immunoelectron , Muscle, Skeletal/ultrastructure
19.
Am J Pathol ; 156(4): 1151-5, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10751338

ABSTRACT

Sporadic inclusion-body myositis (s-IBM) is the most common progressive muscle disease of older persons. Pathologically, the muscle biopsy manifests various degrees of inflammation and specific vacuolar degeneration of muscle fibers characterized by paired helical filaments (PHFs) composed of phosphorylated tau. IBM vacuolated fibers also contain accumulations of several other Alzheimer-characteristic proteins. Molecular mechanisms leading to formation of the PHFs and accumulations of proteins in IBM muscle are not known. We report that the abnormal muscle fibers of IBM contained (i) acridine-orange-positive RNA inclusions that colocalized with the immunoreactivity of phosphorylated tau and (ii) survival motor neuron protein immunoreactive inclusions, which by immuno-electron microscopy were confined to paired helical filaments. This study demonstrates two novel components of the IBM paired helical filaments, which may lead to better understanding of their pathogenesis.


Subject(s)
Muscles/metabolism , Myositis, Inclusion Body/metabolism , Nerve Tissue Proteins/metabolism , Neuropil Threads/metabolism , RNA/metabolism , Acridine Orange , Antibodies, Monoclonal , Cyclic AMP Response Element-Binding Protein , Fluorescent Dyes , Humans , Immunohistochemistry/methods , RNA-Binding Proteins , SMN Complex Proteins , Staining and Labeling , Tissue Distribution
20.
Ann Neurol ; 47(4): 544-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10762172

ABSTRACT

Typical of sporadic inclusion body myositis muscle biopsies are vacuolated muscle fibers containing intracellular amyloid deposits and accumulations of "Alzheimer-characteristic" proteins. There is no muscle blood vessel or cardiac amyloidosis. We report on a 70-year-old African-American man homozygous for the transthyretin Val122Ile allele who has both sporadic inclusion body myositis and cardiac amyloidosis. His unique pathological features included transthyretin immunoreactivity in prominent muscle blood vessel amyloid and congophilic amyloid deposits within vacuolated muscle fibers.


Subject(s)
Amyloidosis/pathology , Inclusion Bodies/pathology , Myocardium/pathology , Myositis/pathology , Point Mutation , Prealbumin/genetics , Aged , Alleles , Biopsy , Coronary Vessels , Homozygote , Humans , Male , Microscopy, Electron , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Myositis/genetics , Neural Conduction , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL