Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Vet Diagn Invest ; 27(6): 762-6, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26450833

ABSTRACT

Chronic inflammatory demyelinating polyradiculoneuropathy occurred in an 11-year-old Labrador Retriever dog. Spinal cord compression resulted from massive radiculitis with prominent cholesterol granulomas. Cholesterol deposition and associated granuloma formation is unique in chronic inflammatory demyelinating polyradiculoneuropathy, in both its human and canine expressions.


Subject(s)
Cholesterol/metabolism , Dog Diseases/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/veterinary , Spinal Nerve Roots/pathology , Animals , Dog Diseases/etiology , Dogs , Female , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/etiology
2.
J Neuropathol Exp Neurol ; 74(7): 688-703, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26083569

ABSTRACT

Autosomal recessive mutations in the RARS2 gene encoding the mitochondrial arginyl-transfer RNA synthetase cause infantile-onset myoencephalopathy pontocerebellar hypoplasia type 6 (PCH6). We describe 2 sisters with novel compound heterozygous RARS2 mutations who presented perinatally with neurologic features typical of PCH6 but with additional features including cardiomyopathy, hydrops, and pulmonary hypoplasia and who died at 1 day and 14 days of age. Magnetic resonance imaging findings included marked cerebellar hypoplasia, gyral immaturity, punctate lesions in cerebral white matter, and unfused deep cerebral grey matter. Enzyme histochemistry of postmortem tissues revealed a near-global cytochrome c oxidase-deficiency; assessment of respiratory chain enzyme activities confirmed severe deficiencies involving complexes I, III, and IV. Molecular genetic studies revealed 2 RARS2 gene mutations: a c.1A>G, p.? variant predicted to abolish the initiator methionine, and a deep intronic c.613-3927C>T variant causing skipping of exons 6-8 in the mature RARS2 transcript. Neuropathologic investigation included low brain weights, small brainstem and cerebellum, deep cerebral white matter pathology, pontine nucleus neuron loss (in 1 sibling), and peripheral nerve pathology. Mitochondrial respiratory chain immunohistochemistry in brain tissues confirmed an absence of complexes I and IV immunoreactivity with sparing of mitochondrial numbers. These cases expand the clinical spectrum of RARS2 mutations, including antenatal features and widespread mitochondrial respiratory chain deficiencies in postmortem brain tissues.


Subject(s)
Aminoacyltransferases/genetics , Cardiomyopathies/genetics , Hydrops Fetalis/genetics , Mitochondrial Diseases/genetics , Mutation/genetics , Olivopontocerebellar Atrophies/genetics , Brain/pathology , Cardiomyopathies/complications , Electron Transport Complex IV/metabolism , Female , Fetus , Humans , Infant, Newborn , Magnetic Resonance Imaging , Mitochondrial Diseases/complications , Molecular Biology , Muscles/pathology , Olivopontocerebellar Atrophies/complications , Postmortem Changes , Pregnancy
3.
J Cell Biol ; 204(7): 1219-36, 2014 Mar 31.
Article in English | MEDLINE | ID: mdl-24687281

ABSTRACT

The PI 3-kinase (PI 3-K) signaling pathway is essential for Schwann cell myelination. Here we have characterized PI 3-K effectors activated during myelination by probing myelinating cultures and developing nerves with an antibody that recognizes phosphorylated substrates for this pathway. We identified a discrete number of phospho-proteins including the S6 ribosomal protein (S6rp), which is down-regulated at the onset of myelination, and N-myc downstream-regulated gene-1 (NDRG1), which is up-regulated strikingly with myelination. We show that type III Neuregulin1 on the axon is the primary activator of S6rp, an effector of mTORC1. In contrast, laminin-2 in the extracellular matrix (ECM), signaling through the α6ß4 integrin and Sgk1 (serum and glucocorticoid-induced kinase 1), drives phosphorylation of NDRG1 in the Cajal bands of the abaxonal compartment. Unexpectedly, mice deficient in α6ß4 integrin signaling or Sgk1 exhibit hypermyelination during development. These results identify functionally and spatially distinct PI 3-K pathways: an early, pro-myelinating pathway driven by axonal Neuregulin1 and a later-acting, laminin-integrin-dependent pathway that negatively regulates myelination.


Subject(s)
Myelin Sheath/physiology , Peripheral Nervous System/cytology , Phosphatidylinositol 3-Kinases/metabolism , Protein Processing, Post-Translational , Animals , Cell Cycle Proteins/metabolism , Cells, Cultured , Coculture Techniques , Extracellular Matrix/metabolism , Gene Expression , Immediate-Early Proteins/metabolism , Integrin beta4/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Laminin/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neuregulin-1/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptors, Laminin/metabolism , Ribosomal Protein S6/metabolism , Schwann Cells/metabolism , Signal Transduction
4.
J Neuropathol Exp Neurol ; 70(5): 386-98, 2011 May.
Article in English | MEDLINE | ID: mdl-21487305

ABSTRACT

We analyzed clinical and pathological disease in 2 peripheral myelin protein-22 (PMP22) overexpressing mouse models for 1.5 years. C22 mice have 7 and C3-PMP mice have 3 to 4 copies of the human PMP22 gene. C3-PMP mice showed no overt clinical signs at 3 weeks and developed mild neuromuscular impairment; C22 mice showed signs at 3 weeks that progressed to severe impairment. Adult C3-PMP mice had very similar, stable, low nerve conduction velocities similar to adults with human Charcot-Marie-Tooth disease type 1A (CMT1A); velocities were much lower in C22 mice. Myelination was delayed, and normal myelination was not reached in either model but the degree of dysmyelination in C3-PMP mice was considerably less than that in C22 mice; myelination was stable in the adult mice. Numbers of myelinated, fibers were reduced at 3 weeks in both models, suggesting that normal numbers of myelinated fibers are not reached during development in the models. In adult C3-PMP and wild-type mice, there was no detectable loss of myelinated fibers,whereas there was clear loss of myelinated fibers in C22 mice.In C3-PMP mice, there is a balance between myelination status and axonal function early in life, whereas in C22 mice, early reduction of axons is more severe and there is major loss of axons in adulthood. We conclude that C3-PMP mice may be an appropriate model for most CMT1A patients, whereas C22 mice may be more relevant to severely affected patients in the CMT1 spectrum.


Subject(s)
Axons/pathology , Demyelinating Diseases/pathology , Myelin Proteins/genetics , Myelin Sheath/pathology , Nerve Fibers, Myelinated/pathology , Neuromuscular Diseases/pathology , Action Potentials/genetics , Animals , Axons/metabolism , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Electrophysiology , Genotype , Mice , Mice, Transgenic , Myelin Proteins/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism , Nerve Fibers, Myelinated/metabolism , Neuromuscular Diseases/genetics , Neuromuscular Diseases/metabolism
6.
Neurobiol Dis ; 42(3): 368-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21303696

ABSTRACT

CMT4D disease is a severe autosomal recessive demyelinating neuropathy with extensive axonal loss leading to early disability, caused by mutations in the N-myc downstream regulated gene 1 (NDRG1). NDRG1 is expressed at particularly high levels in the Schwann cell (SC), but its physiological function(s) are unknown. To help with their understanding, we characterise the phenotype of a new mouse model, stretcher (str), with total Ndrg1 deficiency, in comparison with the hypomorphic Ndrg1 knock-out (KO) mouse. While both models display normal initial myelination and a transition to overt pathology between weeks 3 and 5, the markedly more severe str phenotype suggests that even low Ndrg1 expression results in significant phenotype rescue. Neither model replicates fully the features of CMT4D: although axon damage is present, regenerative capacity is unimpaired and the mice do not display the early severe axonal loss typical of the human disease. The widespread large fibre demyelination coincides precisely with the period of rapid growth of the animals and the dramatic (160-500-fold) increase in myelin volume and length in large fibres. This is followed by stabilisation after week 10, while small fibres remain unaffected. Gene expression profiling of str peripheral nerve reveals non-specific secondary changes at weeks 5 and 10 and preliminary data point to normal proteasomal function. Our findings do not support the proposed roles of NDRG1 in growth arrest, terminal differentiation, gene expression regulation and proteasomal degradation. Impaired SC trafficking failing to meet the considerable demands of nerve growth, emerges as the likely pathogenetic mechanism in NDRG1 deficiency.


Subject(s)
Cell Cycle Proteins/metabolism , Demyelinating Diseases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Myelin Sheath/metabolism , Schwann Cells/metabolism , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Disease Models, Animal , Electrophysiology , Gene Expression , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Myelin Sheath/genetics , Myelin Sheath/pathology , Refsum Disease/genetics , Refsum Disease/metabolism , Refsum Disease/pathology , Schwann Cells/pathology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
7.
Brain ; 134(Pt 2): 618-26, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21278409

ABSTRACT

P. K. Thomas (1926-2008) occupied a prominent place in British and world neurology during the second half of the 20th century. Here, his lasting achievements as clinical neurologist, clinician scientist and experimentalist, editor of monographs and journals and leader of professional developments in the UK and elsewhere are assessed.


Subject(s)
Neurology/history , England , History, 20th Century , History, 21st Century
8.
Neuromuscul Disord ; 19(4): 264-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19272779

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is a heterogeneous group of inherited peripheral motor and sensory neuropathies. The locus responsible for CMT4C was previously assigned to the chromosome 5q23 region by homozygosity mapping and mutations in the SH3TC2 (KIAA1985) gene have been subsequently identified mainly in families around the Mediterranean basin but also frequently in European Gypsies. No English families have been reported to date. To determine the frequency, phenotype and neuropathology of CMT due to SH3TC2 mutations we screened 23 English autosomal recessive (AR) demyelinating CMT families. Five families with AR demyelinating CMT and SH3TC2 mutations were identified, four families were homozygous for the R954X mutation and the fifth family was compound heterozygous for the R954X and E657K mutations. There was significant clinical variation between these families with some cases presenting with a severe childhood onset neuropathy with respiratory and cranial nerve involvement, compared to other families with mild scoliosis and foot deformity. Characteristic sural nerve neuropathology was seen in three families with frequent demyelinating fibres surrounded by excess Schwann cell lamellae forming basal lamina onion bulbs and abnormally long and attenuated Schwann cell processes. One patient homozygous for the R954X mutation had a 20-year history of an inflammatory neuropathy that was superimposed onto the hereditary form, indicating that structural alterations to the SH3TC2 gene could possibly predispose to peripheral nerve inflammation.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Genetic Predisposition to Disease/genetics , Inflammation/genetics , Mutation/genetics , Proteins/genetics , Adult , Age of Onset , Charcot-Marie-Tooth Disease/physiopathology , Child , Chromosome Disorders/genetics , Cranial Nerve Diseases/genetics , DNA Mutational Analysis , Female , Foot Deformities, Congenital/genetics , Genes, Recessive/genetics , Genetic Testing , Genotype , Humans , Inflammation/pathology , Inflammation/physiopathology , Intracellular Signaling Peptides and Proteins , Male , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Phenotype , Respiratory Insufficiency/genetics , Scoliosis/genetics , Sural Nerve/metabolism , Sural Nerve/pathology , Sural Nerve/physiopathology
9.
Eur J Neurosci ; 25(10): 2947-55, 2007 May.
Article in English | MEDLINE | ID: mdl-17561810

ABSTRACT

Phagocytosis is defined as the ingestion of particulates over 0.5 microm in diameter and is associated with cells of the immune system such as macrophages or monocytes. Neurones are not generally recognized to be phagocytic. Using light, confocal, time-lapse and electron microscopy, we carried out a wide range of in-vitro and in-vivo experiments to examine the phagocytic capacity of different neuronal cell types. We demonstrated phagocytosis of material by neurones, including cell debris and synthetic particles up to 2.8 microm in diameter. We showed phagocytosis in different neuronal types, and demonstrated that debris can be transported from neurite extremities to cell bodies and persist within neurones. Flow cytometry analysis demonstrated the lack of certain complement receptors on neurones but the presence of others, including integrin receptors known to mediate macrophage phagocytosis, indicating that a restricted set of phagocytosis receptors may mediate this process. Neuronal phagocytosis occurs in vitro and in vivo, and we propose that this is a more widespread and significant process than previously recognized. Neuronal phagocytosis may explain certain inclusions in neurones during disease, cell-to-cell spread of disease, neuronal death during disease progression and provide a potential mechanism for therapeutic intervention through the delivery of particulate drug carriers.


Subject(s)
Nervous System/metabolism , Neurons/metabolism , Phagocytosis/physiology , Animals , Axons/metabolism , Axons/ultrastructure , Chick Embryo , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Flow Cytometry , Ganglia, Spinal/metabolism , Ganglia, Spinal/ultrastructure , Humans , Integrins/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Electron , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Nervous System/ultrastructure , Neurons/ultrastructure , Rats , Rats, Wistar , Receptors, Cell Surface/metabolism
10.
Ann Neurol ; 60(2): 253-5, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16862591

ABSTRACT

Dysfunction of the ubiquitin-proteasomal system (UPS) has been implicated in the pathogenesis of Parkinson's disease. The systemic administration of UPS inhibitors has been reported to induce nigrostriatal cell death and model Parkinson's disease pathology in rodents. We administered a synthetic, specific UPS inhibitor (PSI) subcutaneously to rats and quantified substantia nigral tyrosine hydroxylase-positive dopaminergic neurons by stereology. PSI caused a 15% decrease in UPS activity at 2 weeks and a 42% reduction in substantia nigra pars compacta tyrosine hydroxylase-positive neurons at 8 weeks. Systemic inhibition of the UPS warrants further evaluation as a means to model Parkinson's disease.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Neurons/enzymology , Oligopeptides/pharmacology , Substantia Nigra/cytology , Substantia Nigra/enzymology , Tyrosine 3-Monooxygenase/metabolism , Animals , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Chymotrypsin/metabolism , Grooming/drug effects , Hand Strength , Motor Activity/drug effects , Muscle Tonus/drug effects , Postural Balance/drug effects , Proteasome Endopeptidase Complex/drug effects , Rats , Rats, Sprague-Dawley , Reflex/drug effects , Weight Gain/drug effects
11.
Neuromuscul Disord ; 16(7): 449-53, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16806930

ABSTRACT

We investigated the manifestations of CMT4C disease in a genetically homogeneous group of patients homozygous for the recently identified Gypsy founder mutation p.Arg1109X in SH3TC2. We observed a surprising degree of variation in age at onset, rate of progression, extent and severity of motor and sensory involvement, scoliosis, and cranial nerve involvement, suggesting that the phenotypic spectrum of CMT4C disease is much broader than the classical diagnostic criteria. Phenotype similarity in first degree relatives and increasing heterogeneity in more distantly related subjects point to the involvement of genetic modifiers, possibly variants in the genes encoding protein partners interacting with SH3TC2.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Point Mutation , Proteins/genetics , Roma/genetics , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Female , Founder Effect , Genotype , Homozygote , Humans , Infant , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Pedigree , Phenotype , Polymorphism, Restriction Fragment Length
12.
Neuromuscul Disord ; 16(2): 126-31, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16427285

ABSTRACT

We present the clinical, imaging and neuropathological findings in three patients with predominant brachial plexus neuropathy. MR scanning was key to determining the brachial plexus involvement. Biopsy of the brachial plexus was performed in one patient. The appearances of the brachial plexus on MRI, in conjunction with the clinical presentations of these patients, suggest that they are unusual variants within the spectrum of chronic inflammatory demyelinating polyradiculoneuropathy.


Subject(s)
Brachial Plexus Neuropathies/diagnosis , Brachial Plexus Neuropathies/pathology , Brachial Plexus/pathology , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/pathology , Adult , Aged , Biopsy , Brachial Plexus/physiopathology , Disease Progression , Female , Humans , Hypertrophy , Magnetic Resonance Imaging , Male , Middle Aged
13.
J Immunol ; 174(4): 1938-46, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699121

ABSTRACT

Multiple sclerosis (MS) is thought to involve CD4 T cell recognition of self myelin, many studies focusing on a pathogenic role for anti-myelin, HLA-DR15-restricted T cells. In experimental allergic encephalomyelitis, it is known which epitopes trigger disease and that disease is associated with determinant spread of T cell reactivity. Characterization of these events in human MS is critical for the development of peptide immunotherapies, but it has been difficult to define the role of determinant spread or define which epitopes might be involved. In this study, we report humanized transgenic mice, strongly expressing HLA-DR15 with an MS-derived TCR; even on a RAG-2 wild-type background, mice spontaneously develop paralysis. Disease, involving demyelination and axonal degeneration, correlates with inter- and intramolecular spread of the T cell response to HLA-DR15-restricted epitopes of myelin basic protein, myelin oligodendrocyte glycoprotein, and alphaB-crystallin. Spread is reproducible and progressive, with two of the epitopes commonly described in responses of HLA-DR15 patients. The fact that this pattern is reiterated as a consequence of CNS tissue damage in mice demonstrates the value of the transgenic model in supplying an in vivo disease context for the human responses. This model, encompassing pathologically relevant, spontaneous disease with the presentation of myelin epitopes in the context of HLA-DR15, should offer new insights and predictions about T cell responses during MS as well as a more stringent test bed for immunotherapies.


Subject(s)
HLA-DR Antigens/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Cell Movement/genetics , Cell Movement/immunology , Central Nervous System/immunology , Central Nervous System/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Disease Progression , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , HLA-DR Antigens/biosynthesis , HLA-DR Antigens/physiology , HLA-DR Serological Subtypes , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Sclerosis/pathology , Myelin Basic Protein/immunology , Myelin Basic Protein/metabolism , Paralysis/genetics , Paralysis/immunology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/physiology , T-Lymphocyte Subsets/pathology
14.
Ann Neurol ; 56(4): 586-90, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15455439

ABSTRACT

There are two known autosomal dominant genes for the hereditary ulcero-mutilating neuropathies: SPTLC1 (hereditary sensory neuropathy type 1) and RAB7 (Charcot-Marie-Tooth disease type 2B). We report a family with autosomal dominant ulcero-mutilating neuropathy, developing in the teens and characterized by ulcers, amputations, sensory involvement in the feet but no motor features. Sequencing the RAB7 gene showed a novel heterozygous A to C mutation, changing asparagine to threonine at codon 161. The mutation is situated adjacent to a previously identified valine to methionine mutation at codon 162, implying a hotspot for mutations in the highly conserved C terminus of RAB7.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation , rab GTP-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Asparagine/genetics , Biopsy/methods , Carrier Proteins/metabolism , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Charcot-Marie-Tooth Disease/physiopathology , DNA Mutational Analysis/methods , Family Health , Functional Laterality/physiology , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/pathology , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Immunohistochemistry/methods , In Vitro Techniques , Male , Middle Aged , Neural Conduction/physiology , Staining and Labeling/methods , Sural Nerve/metabolism , Sural Nerve/pathology , Sural Nerve/physiopathology , Sural Nerve/radiation effects , Threonine/genetics , rab7 GTP-Binding Proteins
15.
Brain ; 127(Pt 1): 193-202, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14607795

ABSTRACT

Classically, the course of Charcot-Marie-Tooth (CMT) disease is gradually progressive. We describe eight atypical patients who developed acute or subacute deterioration. Seven of these had genetically proven CMT disease type 1A (CMT1A) due to chromosome 17p11.2-12 duplication, and one had X-linked disease (CMTX) due to a mutation in the GJB1 gene. In this group there was sufficient clinical, electrophysiological and neuropathological information to indicate a diagnosis of a superimposed inflammatory polyneuropathy. The age range of the patients was 18-69 years, with a mean of 39 years. A family history of a similar neuropathic condition was present in only four patients. All eight had an acute or subacute deterioration following a long asymptomatic or stable period. Seven had neuropathic pain or prominent positive sensory symptoms. Nerve biopsy demonstrated excess lymphocytic infiltration in all eight patients. Five patients were treated with steroids and/or intravenous immunoglobulin, with variable positive response; three patients received no immunomodulatory treatment. Inflammatory neuropathy has previously been recognized in patients with hereditary neuropathy, with uncharacterized genetic defects and with CMT1B. We present detailed assessments of patients with CMT1A and CMTX, including nerve biopsy, and conclude that coexistent inflammatory neuropathy is not genotype-specific in hereditary motor and sensory neuropathy. Although this was not a formal epidemiological study, estimates of the prevalence of CMT disease and chronic inflammatory demyelinating polyneuropathy indicate that the association is more frequent than would be expected by chance. This has implications for understanding the pathogenesis of inflammatory neuropathies and raises important considerations in the management of patients with hereditary neuropathies. If a patient with CMT disease experiences an acute or subacute deterioration in clinical condition, treatment of a coexistent inflammatory neuropathy with steroids or immunoglobulin should be considered.


Subject(s)
Charcot-Marie-Tooth Disease/complications , Guillain-Barre Syndrome/etiology , Acute Disease , Adolescent , Adult , Aged , Biopsy , Charcot-Marie-Tooth Disease/pathology , Charcot-Marie-Tooth Disease/therapy , Chromosomes, Human, Pair 17/genetics , Connexins/genetics , Disease Progression , Female , Glucocorticoids/therapeutic use , Guillain-Barre Syndrome/pathology , Guillain-Barre Syndrome/therapy , Humans , Immunoglobulins, Intravenous , Male , Middle Aged , Neural Conduction , Prednisolone/therapeutic use , Treatment Outcome , Gap Junction beta-1 Protein
16.
Hum Mol Genet ; 12(14): 1713-23, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12837694

ABSTRACT

Charcot-Marie-Tooth disease type 4B1, CMT4B1, is a severe, autosomal-recessive, demyelinating peripheral neuropathy, due to mutations in the Myotubularin-related 2 gene, MTMR2. MTMR2 is widely expressed and encodes a phosphatase whose substrates include phosphoinositides. However, this does not explain how MTMR2 mutants specifically produce demyelination in the peripheral nerve. Therefore, we analysed the cellular and subcellular distribution of Mtmr2 in nerve. Mtmr2 was detected in all cytoplasmic compartments of myelin-forming Schwann cells, as well as in the cytoplasm of non-myelin-forming Schwann cells and both sensory and motorneurons. In contrast, Mtmr2 was detected in the nucleus of Schwann cells and motorneurons, but not in the nucleus of sensory neurons. As Mtmr2 is diffusely present also within the nerve, a specific function could derive instead from nerve-specific interacting proteins. Therefore, we performed two yeast two-hybrid screenings, using either fetal brain or peripheral nerve cDNA libraries. The neurofilament light chain protein, NF-L, was identified repeatedly in both screenings, and found to interact with MTMR2 in both Schwann cells and neurons. Interestingly, NF-L, encoding NF-L, is mutated in CMT2E. These data may provide a basis for the nerve-specific pathogenesis of CMT4B1, and further support for the notion that hereditary demyelinating and axonal neuropathies may represent different clinical manifestations of a common pathological mechanism.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Neurofilament Proteins/metabolism , Peripheral Nerves/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Charcot-Marie-Tooth Disease/metabolism , Humans , Mice , Mutation , Neurofilament Proteins/genetics , Neurons/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor , Rats , Schwann Cells/metabolism
17.
J Peripher Nerv Syst ; 8(1): 13-6, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12678549

ABSTRACT

Chronic inflammatory demyelinating polyneuropathy (CIDP) may cause significant disability, but severe respiratory complications are uncommon. We describe the case of a 49-year-old man with clinical features of CIDP for 5 years who died of respiratory failure. Post-mortem findings of denervation in diaphragm muscle and axonal loss in phrenic nerve are presented. Severe ventilatory failure may occur in CIDP when neuropathy affects the respiratory muscles. Attention to early clinical features of respiratory insufficiency may facilitate the prevention of more severe features of respiratory failure.


Subject(s)
Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/pathology , Respiratory Insufficiency/pathology , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Sural Nerve/pathology
18.
Hum Mol Genet ; 11(9): 1075-94, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11978767

ABSTRACT

Machado-Joseph disease (MJD; MIM 109150) is a late-onset neurodegenerative disorder caused by the expansion of a polyglutamine tract within the MJD1 gene. We have previously reported the generation of human yeast artificial chromosome (YAC) constructs encompassing the MJD1 locus into which expanded (CAG)(76) and (CAG)(84) repeat motifs have been introduced by homologous recombination. Transgenic mice containing pathological alleles with polyglutamine tract lengths of 64, 67, 72, 76 and 84 repeats, as well as the wild type with 15 repeats, have now been generated using these YAC constructs. The mice with expanded alleles demonstrate a mild and slowly progressive cerebellar deficit, manifesting as early as 4 weeks of age. As the disease progresses, pelvic elevation becomes markedly flattened, accompanied by hypotonia, and motor and sensory loss. Neuronal intranuclear inclusion (NII) formation and cell loss is prominent in the pontine and dentate nuclei, with variable cell loss in other regions of the cerebellum from 4 weeks of age. Interestingly, peripheral nerve demyelination and axonal loss is detected in symptomatic mice from 26 weeks of age. In contrast, transgenic mice carrying the wild-type (CAG)(15) allele of the MJD1 locus appear completely normal at 20 months. Disease severity increases with the level of expression of the expanded protein and the size of the repeat. These mice are representative of MJD and will be a valuable resource for the detailed analysis of the roles of repeat length, tissue specificity and level of expression in the neurodegenerative processes underlying MJD pathogenesis.


Subject(s)
Cerebellum/pathology , Machado-Joseph Disease/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Peptides/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Ataxin-3 , Blotting, Western , Cerebellum/metabolism , Chromosomes, Artificial, Yeast/genetics , DNA Primers/chemistry , Demyelinating Diseases/pathology , Female , Humans , Immunoenzyme Techniques , Machado-Joseph Disease/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Mutation/genetics , Nuclear Proteins , Polymerase Chain Reaction , RNA, Messenger/metabolism , Repressor Proteins , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...