Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
IEEE Trans Biomed Circuits Syst ; 3(2): 108-16, 2009 Apr.
Article in English | MEDLINE | ID: mdl-23853202

ABSTRACT

Tremor, the most common form of movement disorder, is an often debilitating condition that adversely affects an individual's ability to maintain functional independence. Efforts to study, diagnose, and treat such movement disorders are complicated by a dearth of quantitative, precise, or accurate methods for motion data collection and assessment. To address this deficiency, this paper provides two contributions: 1) the design of a body-area inertial sensing system and 2) the evaluation of postcapture, on-body signal-processing algorithms that transform sensed inertial data into clinically significant information pertaining to tremor symmetry. For the former, we present our technology that meets requirements for wearability, fidelity, battery life, and interoperability. For the latter, we demonstrate the efficacy of using filter-bank analysis and cross correlation to interpret tremor frequency and energy. We extend the previous work by presenting a wireless body-area inertial sensing technology and a method to reduce, by up to 30 times, the computational demands of cross correlation on such a resource-constrained technology. These efforts lay the foundation for real-time, on-body assessment of tremor as well as more intelligent and energy-efficient data transmission and storage decisions.

2.
Neuroscience ; 133(2): 543-53, 2005.
Article in English | MEDLINE | ID: mdl-15885922

ABSTRACT

Current work emphasizes that peripheral tissue injury and inflammation results in a heightened sensitivity to subsequent noxious input (hyperalgesia) that is mediated in large part by the spinal synthesis and release of eicosanoids, in particular prostaglandins. Secreted phospholipase A(2)s (sPLA(2)s) form a class of structurally related enzymes that release arachidonic acid from cell membranes that is further processed to produce eicosanoids. We hypothesized that spinal sPLA(2)s may contribute to inflammation-induced hyperalgesia. Spinal cord tissue and cerebrospinal fluid were collected from rats for assessment of sPLA(2) protein expression and sPLA(2) activity. A basal sPLA(2) protein expression and activity was detected in spinal cord homogenate (87+/-17 pmol/min/mg), though no activity could be detected in cisternal cerebrospinal fluid, of naive rats. The sPLA(2) activity did not change in spinal cord tissue or cerebrospinal fluid assessed over 8 h after injection of carrageenan into the hind paw. However, the sPLA(2) activity observed in spinal cord homogenates was suppressed by addition of LY311727, a selective sPLA(2) inhibitor. To determine the role of this spinal sPLA(2) in hyperalgesia, we assessed the effects of lumbar intrathecal (IT) administration of LY311727 in rats with chronic IT catheters in three experimental models of hyperalgesia. IT LY311727 (3-30 microg) dose-dependently prevented intraplantar carrageenan-induced thermal hyperalgesia and formalin-induced flinching, at doses that had no effect on motor function. IT LY311727 also suppressed thermal hyperalgesia induced by IT injection of substance P (30 nmol). Using in vivo spinal microdialysis, we found that IT injection of LY311727 attenuated prostaglandin E(2) release into spinal dialysate otherwise evoked by the IT injection of substance P. Taken together, this work points to a role for constitutive sPLA(2)s in spinal nociceptive processing.


Subject(s)
Hyperalgesia/enzymology , Inflammation/physiopathology , Phospholipases A/metabolism , Spinal Cord/enzymology , Animals , Blotting, Western/methods , Carrageenan , Dialysis/methods , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Functional Laterality , Gene Expression Regulation, Enzymologic/drug effects , Group II Phospholipases A2 , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Indoles/pharmacology , Inflammation/chemically induced , Inflammation/complications , Inflammation/enzymology , Inhibition, Psychological , Male , Pain Measurement/drug effects , Pain Measurement/methods , Phospholipases A2 , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Spinal Cord/drug effects , Substance P/pharmacology , Time Factors
3.
J Neurol Sci ; 189(1-2): 37-43, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11535231

ABSTRACT

We report laminin alpha 2 (merosin) deficiency associated with muscular dystrophy and demyelinating neuropathy in two cats. The cats developed progressive muscle weakness, and atrophy. Either hypotonia or contractures resulted in recumbency, necessitating euthanasia. Muscle biopsies showed dystrophic changes including marked endomysial fibrosis, myofiber necrosis, variability of fiber size, and perimysial lipid accumulation. Immunohistochemistry showed that laminin alpha 2 chain was absent or reduced, while dystrophin and all the components of the dystrophin-associated glycoprotein complex were present and normal. One cat was examined in detail. Motor nerve conduction velocity (MNCV) was decreased, and ultrastructurally the peripheral nerves showed Schwann cell degeneration and demyelination. Brain imaging was not performed, but white matter changes were not apparent in the brain at necropsy. The disease in these cats is similar to primary or secondary merosin (laminin alpha 2)-deficient congenital muscular dystrophy (CMD) in humans and to dystrophia muscularis in mice.


Subject(s)
Cat Diseases/genetics , Demyelinating Diseases/veterinary , Laminin/deficiency , Muscle Proteins/deficiency , Muscular Dystrophy, Animal/genetics , Nerve Tissue Proteins/deficiency , Animals , Cat Diseases/pathology , Cats , Contracture/etiology , Demyelinating Diseases/complications , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Female , Laminin/genetics , Muscle Proteins/analysis , Muscle Proteins/genetics , Muscle Rigidity/etiology , Muscle, Skeletal/chemistry , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Dystrophy, Animal/complications , Muscular Dystrophy, Animal/pathology , Nerve Tissue Proteins/genetics , Neural Conduction , Peroneal Nerve/chemistry , Peroneal Nerve/pathology
4.
Br J Haematol ; 108(3): 641-52, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10759724

ABSTRACT

Vectors encoding immunostimulatory genes are under investigation for their use as adjuvants for immunotherapy. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a prominent candidate gene for this approach because this cytokine can prime immune responses to 'self' tumour or other weak antigens. Prior studies suggested that GM-CSF induces accumulation and differentiation of antigen-presenting cells, particularly dendritic cells that can initiate immunity. To evaluate this model in vivo, we performed i.m. and i.p. injections of an adenovirus vector encoding murine GM-CSF (Ad-mGM-CSF) and evaluated local and systemic effects. After intramuscular injection, local changes were characterized by the accumulation of myeloid cells, a subsequent infiltration of lymphocytes and then myonecrosis. Intraperitoneal injection also induced an accumulation of myeloid cells, an increase in CD3-positive T and a decrease in B220-positive B lymphocytes. Expression of the dendritic cell marker CD11c on 48 +/- 9% of the peritoneal cells (n = 6) along with high levels of surface MHC class II, a characteristic morphology, and endocytosis of FITC-dextran suggested in vivo differentiation of dendritic cells after i.p. injection of Ad-mGM-CSF. Systemic effects were observed after i.m. and i.p. injection of Ad-mGM-CSF. All mice developed hepatosplenomegaly resulting from extramedullary haematopoiesis. These changes were specific to GM-CSF as they were not seen in mice injected with an adenovirus vector without a transgene. Our observations indicate that adenoviral transfer of GM-CSF is a powerful tool for inducing local and systemic expansion of haematopoietic cells. The local expansion of myeloid cells displaying signs of dendritic cell differentiation, as characterized for the peritoneal cell compartment, can explain the potency of GM-CSF when used as an adjuvant in genetic immunotherapy.


Subject(s)
Adenoviridae/genetics , Dendritic Cells/cytology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Stem Cells/cytology , Animals , Cell Differentiation/immunology , Cell Division/immunology , Dendritic Cells/ultrastructure , Female , Gene Expression , HeLa Cells , Hematopoiesis, Extramedullary , Humans , Injections, Intramuscular , Injections, Intraperitoneal , Leukocytes/immunology , Liver/anatomy & histology , Liver/physiology , Mice , Mice, Inbred BALB C , Microscopy, Electron , Muscle, Skeletal/anatomy & histology , Organ Size , Spleen/anatomy & histology , Spleen/physiology , Stem Cells/ultrastructure
5.
Ultrastruct Pathol ; 23(5): 285-97, 1999.
Article in English | MEDLINE | ID: mdl-10582266

ABSTRACT

Crystalloid inclusions or "pole bodies" observed in brain macrophages in human demyelinating disease represent a morphological enigma. Similar inclusions were detected in brain macrophages from the GFAP-IL3 mouse, a transgenic murine model for macrophage mediated demyelination. Mice also showed inclusions in hematopoietic tissue. They appear to be related to phagocytosis and secretion, respectively, as evidenced by the fact that in phagocytosing cells they often merged with lysozomes and that affected cells showed empty channels open to the interstitium. Based on ultrastructural and immunolocalization studies using chaperonin-10, lysozyme, and cathepsin the authors suggest that these inclusions are consistent with phagocytosis-related secretory products. This study may provide insight into the nature and significance of similar macrophage inclusions recently identified in multiple sclerosis.


Subject(s)
Bone Marrow Cells/ultrastructure , Cerebellum/pathology , Glial Fibrillary Acidic Protein/biosynthesis , Inclusion Bodies/ultrastructure , Interleukin-3/biosynthesis , Macrophages/ultrastructure , Multiple Sclerosis/pathology , Animals , Bone Marrow Cells/metabolism , Cathepsins/metabolism , Cerebellum/metabolism , Chaperonin 10/metabolism , Crystallization , Disease Models, Animal , Glial Fibrillary Acidic Protein/genetics , Inclusion Bodies/metabolism , Interleukin-3/genetics , Macrophages/metabolism , Mice , Mice, Transgenic , Multiple Sclerosis/etiology , Multiple Sclerosis/metabolism , Muramidase/metabolism
6.
Brain Res ; 835(1): 46-61, 1999 Jul 17.
Article in English | MEDLINE | ID: mdl-10448195

ABSTRACT

Cytokines belonging to the type I interferon (e.g. interferon-alpha) family are important in the host response to infection and may have complex and broad ranging actions in the central nervous system (CNS) that may be beneficial or harmful. To better understand the impact of the CNS expression of the type I interferons (IFN), transgenic mice were developed that produce IFN-alpha(1) chronically from astrocytes. In two independent transgenic lines with moderate and low levels of astrocyte IFN-alpha mRNA expression respectively, a spectrum of transgene dose- and age-dependent structural and functional neurological alterations are induced. Structural changes include neurodegeneration with loss of cholinergic neurons, gliosis, angiopathy with mononuclear cell cuffing, progressive calcification affecting basal ganglia and cerebellum and the up-regulation of a number of IFN-alpha-regulated genes. At a functional level, in vivo and in vitro electrophysiological studies revealed impaired neuronal function and disturbed synaptic plasticity with pronounced hippocampal hyperexcitability. Severe behavioral alterations were also evident in higher expressor GFAP-IFNalpha mice which developed fatal seizures around 13 weeks of age precluding their further behavioral assessment. Modest impairments in discrimination learning were measured in lower expressor GFAP-IFNalpha mice at various ages (7-42 weeks). The behavioral and electrophysiological findings suggest regional changes in hippocampal excitability which may be linked to abnormal calcium metabolism and loss of cholinergic neurons in the GIFN mice. Thus, these transgenic mice provide a novel animal model in which to further evaluate the mechanisms that underlie the diverse actions of type I interferons in the intact CNS and to link specific structural changes with functional impairments.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/pathology , Interferon-alpha/biosynthesis , Nervous System Diseases/genetics , Animals , Behavior, Animal/physiology , Electrophysiology , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Interferon-alpha/genetics , Mice , Mice, Transgenic , Nervous System Diseases/metabolism , Nervous System Diseases/pathology
7.
Hear Res ; 131(1-2): 22-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10355601

ABSTRACT

The MRL-Fas(lpr) mouse, a model of multisystemic, organ nonspecific autoimmune disease, has been proposed as a model of immune-mediated inner ear disease. A preliminary study employing light microscopy indicated that it develops cochlear pathology that appeared most striking in the stria vascularis, where cells underwent edema and degeneration. However, other structures, including the inner and outer hair cells and the supporting cells, also appeared to display pathology. The current study analyzed cochlear ultrastructure using transmission electron microscopy to better delineate the cochlear lesions found in these animals. MRL-Fas(lpr) animals were allowed to develop systemic disease (20 weeks old) and then had auditory brainstem response (ABR) thresholds determined. Animals were then killed and their cochleas prepared for electron microscopy. Age-matched MRL-+/+ and BALB/c mice served as controls. Results indicated that MRL-Fas(lpr) mice demonstrated elevated ABR thresholds. In contrast to a preliminary report, the cochlear pathology was observed exclusively in the stria vascularis, where cells demonstrated hydropic degeneration. Strial capillary structure was normal as were the rest of the cellular cochlear constituents. No inflammatory infiltrate was noted. These studies confirm that the MRL-Fas(lpr) mouse develops cochlear abnormalities focused in the stria vascularis. Whether the mechanism of the cellular degeneration involves autoimmune, genetic, or uremic processes has yet to be determined.


Subject(s)
Mice, Inbred MRL lpr/anatomy & histology , Stria Vascularis/ultrastructure , Animals , Audiometry , Evoked Potentials, Auditory, Brain Stem/physiology , Female , Mice , Mice, Inbred MRL lpr/physiology , Microscopy, Electron , Reference Values
8.
Brain Pathol ; 9(2): 219-35, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10219739

ABSTRACT

Transgenic mice overexpressing cytokines facilitate analysis of the effects of these immunomodulators on indigenous cells of the central nervous system. This study examines morphological aspects of demyelination and permeability changes, in a recently described transgenic model (termed GFAP-IL3). GFAP-IL3 mice develop progressive motor disease at approximately 5 months. Lesions identified after disease onset, showed activation of microglia, astroglial proliferation with phagocytosis of lipids, and immigration of macrophages and mast cells into neural parenchyma. Lymphocytes failed to appear until the later stages of the disease. Later, cerebellar and brain stem white matter contained focal demyelinating lesions with intense macrophage infiltration and a proliferative astrocytosis. Dystrophic axonal changes were noted, in addition to demyelination in heavily infiltrated lesions. Mast cells, variably present in the thalamus and meninges of wild type mice, were greatly increased at these sites in GFAP-IL3 mice. Blood-brain barrier (BBB) defects were documented with leakage of intravenously injected horseradish peroxidase. Mast cell infiltration into the CNS and their degranulation at the site of injury, may represent initial events in a spontaneous process of macrophage mediated demyelination in which glial cells and macrophages are both involved in the phagocytic process.


Subject(s)
Astrocytes/pathology , Blood-Brain Barrier , Brain/pathology , Demyelinating Diseases/pathology , Interleukin-3/genetics , Mast Cells/pathology , Neuroglia/pathology , Animals , Astrocytes/immunology , Astrocytes/ultrastructure , Axonal Transport , Axons/pathology , Axons/ultrastructure , Brain/physiopathology , Cell Division , Cerebellum/pathology , Cerebellum/ultrastructure , Demyelinating Diseases/genetics , Demyelinating Diseases/physiopathology , Glial Fibrillary Acidic Protein/analysis , Glial Fibrillary Acidic Protein/biosynthesis , Horseradish Peroxidase , Interleukin-3/analysis , Interleukin-3/physiology , Mast Cells/physiology , Mice , Mice, Transgenic , Neuroglia/physiology , Neuroglia/ultrastructure , Organ Specificity , Phagocytosis
9.
J Peripher Nerv Syst ; 4(1): 19-26, 1999.
Article in English | MEDLINE | ID: mdl-10197062

ABSTRACT

Multiple Renaut bodies were identified by light microscopy in the biopsied common peroneal nerve of a dog with generalized neuropathy, hypothyroidism and a history of cutaneous mastocytosis. In addition, numerous granulated cells were associated with most of the Renaut bodies. Electron microscopic examination confirmed these to be mast cells, both central and peripheral to Renaut bodies, a phenomenon never previously reported. Endoneurial fibrosis, myelinated fiber loss, as well as evidence of axonal degeneration, demyelination and remyelination were observed. 'Vacuolation' of endoneurial fibroblasts was also present. The location of these Renaut bodies in the common peroneal nerve, and the absence of any documented or expected nerve compression, implicates other etiological factors. These observations are the first to report an association between mast cells and Renaut bodies. It is possible that mast cells, at least in this case, are involved in the formation of Renaut bodies.


Subject(s)
Dog Diseases/immunology , Inclusion Bodies/immunology , Mastocytosis/veterinary , Peripheral Nervous System Diseases/veterinary , Peroneal Nerve/pathology , Animals , Biopsy , Dog Diseases/pathology , Dogs , Fibroblasts/pathology , Fibroblasts/ultrastructure , Fibrosis , Hypothyroidism/immunology , Hypothyroidism/pathology , Hypothyroidism/veterinary , Inclusion Bodies/pathology , Inclusion Bodies/ultrastructure , Male , Mast Cells/immunology , Mast Cells/ultrastructure , Mastocytosis/immunology , Mastocytosis/pathology , Microscopy, Electron , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/pathology , Peroneal Nerve/immunology
10.
J Glaucoma ; 8(1): 38-45, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10084273

ABSTRACT

PURPOSE: To assess relative changes in different tissue components of optic nerve and their relationship to nerve fiber loss in the experimental monkey model of glaucoma. METHODS: Chronic intraocular pressure (IOP) elevation was induced by laser trabeculoplasty in the right eye of eight monkeys (Macaca fascicularis). Both experimental right optic nerves and control left optic nerves were studied. Histomorphometric analysis was performed on optic nerve cross-sections using bright field microscopy with camera lucida. Cross-sectional areas of optic nerve tissue components were estimated by point counting. Nerve fiber density was estimated by unbiased random sampling. Nerve fiber number was calculated by multiplying nerve fiber density with neuroglial area. RESULTS: Varying degrees of nerve fiber loss were seen in eight optic nerves with chronic IOP elevation. More than 50% nerve fiber loss was noted in four of eight experimental optic nerves. In these severely affected optic nerves, total optic nerve area was significantly decreased compared with control optic nerves. Among the optic nerve tissue components, only the ratio of myelinated fiber area to total optic nerve area was significantly decreased. The ratio of extraaxonal area to total optic nerve area was significantly increased, whereas the ratio of interfascicular septal area to total optic nerve area did not change significantly. For all optic nerves, differences in nerve fiber count between control and experimental optic nerves showed the strongest correlation with differences in myelinated fiber area, followed by differences in extraaxonal area and total optic nerve area. CONCLUSION: This histomorphometric study suggests the validity of the experimental monkey model of glaucoma in studying changes occurring in the nonaxonal optic nerve tissue components in human glaucomatous optic neuropathy. Glial scar tissue area was significantly increased in optic nerves with severe glaucomatous damage. Although a decrease in total optic nerve area was observed, among the optic nerve tissue components only myelinated nerve fiber area decreased significantly. Myelinated nerve fiber area also showed the strongest association with nerve fiber loss in experimental glaucoma.


Subject(s)
Glaucoma/pathology , Optic Nerve/pathology , Animals , Cell Count , Chronic Disease , Disease Models, Animal , Follow-Up Studies , Glaucoma/complications , Intraocular Pressure , Macaca fascicularis , Nerve Fibers, Myelinated/pathology , Neuroglia/pathology , Optic Atrophy/etiology , Optic Atrophy/pathology
11.
Muscle Nerve ; 22(2): 156-65, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10024128

ABSTRACT

A transgenic mouse model has been established in which the cytokine interferon-gamma (IFN-gamma) is overexpressed through the action of the acetylcholine receptor epsilon promoter acting at the neuromuscular junction. While originally developed as a model for the study of the pathogenesis of myasthenia gravis, there are important differences from both human myasthenia gravis and its animal model, experimental autoimmune myasthenia gravis. By 4 months of age there was a well-established inflammatory, predominantly necrotizing myopathy, with marked dystrophic calcification. Dystrophic and degenerative changes in terminal axons and adjacent Schwann cells were also apparent. The acetylcholine receptor was not the primary target of the inflammatory response, since at 10 weeks of age the receptor content was not decreased and antibodies were not detected bound to the receptor. The IFNgamma transgenic mouse model may provide a clinically relevant model of necrotizing myopathy for investigation of the pathological changes associated with, and presumably precipitated by, overexpression of the pro-inflammatory cytokine interferon-gamma on the neuromuscular junction, intramuscular nerves and myofibers.


Subject(s)
Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Muscle, Skeletal/pathology , Neuromuscular Diseases/genetics , Neuromuscular Diseases/pathology , Animals , Animals, Newborn , Antibodies/metabolism , Calcinosis/pathology , Disease Models, Animal , Histocytochemistry , Immunohistochemistry , Inflammation/pathology , Mice , Mice, Transgenic , Microscopy, Electron , Motor Endplate/pathology , Motor Endplate/ultrastructure , Motor Neurons/pathology , Motor Neurons/ultrastructure , Muscle, Skeletal/ultrastructure , Necrosis , Receptors, Cholinergic/analysis , Receptors, Cholinergic/metabolism
12.
J Vet Intern Med ; 12(6): 424-30, 1998.
Article in English | MEDLINE | ID: mdl-9857334

ABSTRACT

The diagnosis of nemaline rod myopathy (NM) is based on the presence of numerous pathognomonic rods within a fresh frozen muscle biopsy specimen. Three forms of congenital NM have been described in humans, and rods have been found to occur in various other conditions. A similar myopathy was described in 1986 in a family of cats. In this report, we describe a case of congenital NM in a 10-month-old Border Collie, an adult-onset NM in an 11-year-old Schipperke, and 2 acquired myopathies with nemaline rods in adult dogs associated with hypothyroidism and Cushing's syndrome. Common clinical features included exercise intolerance, abnormal electromyography, and the presence of nemaline rods in fresh, frozen, and glutaraldehyde-fixed biopsies from proximal appendicular limb muscles. Staining of cryostat sections of muscle biopsy specimens by the modified Gomori trichrome technique disclosed numerous rod bodies that were localized to type 1 fibers by the histochemical adenosine triphosphatase reaction. Accumulation of rods also was demonstrated by electron microscopy in 2 of the cases with localized enlargement and streaming of Z lines. Documentation of NM in a young Border Collie and the adult-onset form in the Schipperke alerts clinicians to the existence of this disorder in these breeds.


Subject(s)
Dog Diseases/diagnosis , Myopathies, Nemaline/veterinary , Animals , Azo Compounds/chemistry , Biopsy/veterinary , Coloring Agents/chemistry , Diagnosis, Differential , Dog Diseases/physiopathology , Dogs , Electromyography/veterinary , Eosine Yellowish-(YS)/chemistry , Female , Hypertrophy/diagnosis , Hypertrophy/physiopathology , Hypertrophy/veterinary , Male , Methyl Green/chemistry , Microscopy, Electron , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Myopathies, Nemaline/diagnosis , Myopathies, Nemaline/physiopathology
13.
Am J Pathol ; 153(3): 767-83, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9736027

ABSTRACT

To examine the role of tumor necrosis factor (TNF)-alpha in the pathogenesis of degenerative disorders of the central nervous system (CNS), transgenic mice were developed in which expression of murine TNF-alpha was targeted to astrocytes using a glial fibrillary acidic protein (GFAP)-TNF-alpha fusion gene. In two independent GFAP-TNFalpha transgenic lines (termed GT-8 or GT-2) adult (>4 months of age) animals developed a progressive ataxia (GT-8) or total paralysis affecting the lower body (GT-2). Symptomatic mice had prominent meningoencephalitis (GT-8) or encephalomyelitis (GT-2) in which large numbers of B cells and CD4+ and CD8+ T cells accumulated at predominantly perivascular sites. The majority of these lymphocytes displayed a memory cell phenotype (CD44high, CD62Llow, CD25-) and expressed an early activation marker (CD69). Parenchymal lesions contained mostly CD45+ high, MHC class II+, and Mac-1+ cells of the macrophage microglial lineage with lower numbers of neutrophils and few CD4+ and CD8+ T cells. Cerebral expression of the cellular adhesion molecules ICAM-1, VCAM-1, and MAdCAM as well as a number of alpha- and beta-chemokines was induced or upregulated and preceded the development of inflammation, suggesting an important signaling role for these molecules in the CNS leukocyte migration. Degenerative changes in the CNS of the GFAP-TNFalpha mice paralleled the development of the inflammatory lesions and included primary and secondary demyelination and neurodegeneration. Disease exacerbation with more extensive inflammatory lesions that contained activated cells of the macrophage/microglial lineage occurred in GFAP-TNFalpha mice with severe combined immune deficiency. Thus, persistent astrocyte expression of murine TNF-alpha in the CNS induces a late-onset chronic inflammatory encephalopathy in which macrophage/microglial cells but not lymphocytes play a central role in mediating injury.


Subject(s)
Astrocytes/metabolism , Encephalomyelitis/etiology , Immunocompetence , Meningoencephalitis/etiology , Mice, SCID/metabolism , Severe Combined Immunodeficiency/complications , Tumor Necrosis Factor-alpha/physiology , Animals , Astrocytes/pathology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Brain/metabolism , Brain/pathology , Cell Adhesion Molecules/metabolism , Encephalomyelitis/metabolism , Encephalomyelitis/pathology , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Immunophenotyping , In Situ Hybridization , Male , Meningoencephalitis/metabolism , Meningoencephalitis/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Fusion Proteins/metabolism , Severe Combined Immunodeficiency/metabolism , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
14.
J Neuropathol Exp Neurol ; 57(3): 268-82, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9600219

ABSTRACT

Transgenic mice, named GFAP-IL6, that express interleukin-6 in astrocytes in the central nervous system (CNS) have a constitutive blood-brain barrier (BBB) defect and develop a progressive neurodegenerative disease. Based on ultrastructural observations showing electron-dense pigment in the brain of the GFAP-IL6 mice, we hypothesized that iron metabolism was altered in the brains of these animals. Enhanced histochemical methods revealed abnormal iron deposition in the cerebellum from 1 month of age that worsened with progression of the disease. Immunohistochemical analysis of iron-binding proteins (IBP) showed increased ferritin immunoreactivity and a decreased signal from the transferrin receptor in symptomatic animals. Atomic absorption spectroscopy revealed a 40% increase of total iron concentration in the cerebellum at the symptomatic stage. In order to obtain evidence that accumulation of this oxidizing metal was toxic, we looked for the presence of oxidative damage. Using the MAL-2 antibody, extensive lipid peroxidation (LP) was detected in the neocortex and the cerebellum in symptomatic animals. Ultrastructural analysis indicated lipofuscin deposition at the sites of neuro-axonal degeneration and abnormal iron deposition. These results suggest that the IL6-induced BBB defect precipitates iron accumulation in the GFAP-IL6 mouse brain and that subsequent IBP regulation mediates protective responses. As these defenses become overwhelmed, the iron overload seems to promote LP, which may contribute to the neurodegeneration that ensues. This transgenic mouse model of IL6-mediated neurodegeneration provides a unique opportunity to examine several aspects of iron metabolism in the brain, including its entry at the site of the BBB, its distribution through the IBP, and its mechanisms of toxicity.


Subject(s)
Brain/metabolism , Interleukin-6/metabolism , Iron/metabolism , Lipid Peroxidation , Animals , Blood-Brain Barrier/physiology , Brain/pathology , Carrier Proteins/metabolism , Female , Ferritins/blood , Glial Fibrillary Acidic Protein/genetics , Immunohistochemistry , Interleukin-6/genetics , Iron-Binding Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Receptors, Transferrin/metabolism , Spectrophotometry, Atomic , Transferrin/metabolism , Transferrin-Binding Proteins
15.
Arch Ophthalmol ; 116(2): 178-85, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9488269

ABSTRACT

OBJECTIVES: To determine the incidence of clinical resistance to intraocular cidofovir injection for treatment of acquired immunodeficiency syndrome (AIDS)-related cytomegalovirus (CMV) retinitis, and to identify virologic features associated with cidofovir treatment failure. PATIENTS AND METHODS: Clinical resistance to intravitreal cidofovir was examined in 64 patients with CMV retinitis who received at least 1 injection of 20 pg of cidofovir. Histopathologic examination, culture, and polymerase chain reaction were used to detect CMV in ocular specimens. Antiviral resistance was assessed by plaque reduction assay and DNA sequencing. RESULTS: Clinical resistance to intravitreal cidofovir injections was identified in 3 patients (5%) and was associated with prior oral ganciclovir or intravenous cidofovir use. Ganciclovir- and cidofovir-resistant CMV isolates were cultured from 2 patients and harbored resistance-associated mutations in the UL97 and polymerase genes. Resistance mutations were also detected by direct analysis of vitreous. In 1 patient, different resistance mutations were identified in ocular vs extraocular CMV strains. CONCLUSIONS: Clinical failure of intravitreal cidofovir occurs infrequently, but may be associated with cidofovir-resistant CMV selected by prior ganciclovir or cidofovir treatment. Ocular CMV disease can result from a localized infection with a resistant CMV strain, and antiviral resistance may develop at a local site of infection independently from resistance that develops systemically.


Subject(s)
AIDS-Related Opportunistic Infections/drug therapy , Antiviral Agents/therapeutic use , Cytomegalovirus Retinitis/drug therapy , Cytomegalovirus/drug effects , Cytosine/analogs & derivatives , Organophosphonates , Organophosphorus Compounds/therapeutic use , AIDS-Related Opportunistic Infections/pathology , AIDS-Related Opportunistic Infections/virology , Adult , Cidofovir , Cytomegalovirus/genetics , Cytomegalovirus/isolation & purification , Cytomegalovirus Retinitis/pathology , Cytomegalovirus Retinitis/virology , Cytosine/therapeutic use , DNA, Viral/analysis , Drug Resistance, Microbial , Ganciclovir/therapeutic use , Humans , Injections , Male , Middle Aged , Polymerase Chain Reaction , Treatment Failure , Vitreous Body
16.
Acta Neuropathol ; 95(2): 171-4, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9498053

ABSTRACT

Nerve biopsy samples from two cats with spontaneously occurring diabetes were examined. The predominant nerve fiber abnormalities observed were restricted to the myelin sheath and Schwann cell. Reactive, degenerative and proliferative Schwann cell changes were evident but the most striking abnormality encountered was splitting and ballooning of the myelin sheath. These observations highlight the significance of Schwann cell injury in the pathogenesis of diabetic neuropathy.


Subject(s)
Cat Diseases , Demyelinating Diseases/veterinary , Diabetes Mellitus, Type 1/veterinary , Diabetic Neuropathies/veterinary , Myelin Sheath/pathology , Schwann Cells/pathology , Animals , Cats , Cell Division , Demyelinating Diseases/etiology , Demyelinating Diseases/pathology , Diabetes Mellitus, Type 1/pathology , Diabetic Neuropathies/complications , Diabetic Neuropathies/pathology , Female , Male , Myelin Sheath/ultrastructure , Nerve Fibers/pathology , Nerve Fibers/ultrastructure , Neural Conduction , Orchiectomy , Ovariectomy , Schwann Cells/ultrastructure
17.
Acta Neuropathol ; 95(1): 47-56, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9452821

ABSTRACT

Despite early descriptions of hypertrophic Schwann cells and onion-bulb formation in patients with diabetic neuropathy, clinical and experimental studies have emphasized axonal pathology. In recent years, the Schwann cell has been further implicated in diabetic neuropathy because it is the primary intrafascicular location for the first enzyme of the polyol pathway, aldose reductase, which appears to have a role in modulating a variety of complications of diabetes, including diabetic neuropathy. To further explore the role of polyol pathway flux in the pathogenesis of Schwann cell injury, ultrastructural abnormalities of Schwann cells in human diabetic neuropathy (HDN) were compared with those in experimental galactose neuropathy (EGN), a well-characterized model of hyperglycemia without hypoinsulinemia. Similar to previous studies of EGN, reactive, degenerative and proliferative changes of Schwann cells were observed after 2, 4 and 24 months of galactose intoxication. Reactive changes included accumulation of lipid droplets, pi granules of Reich and glycogen granules, increased numbers of subplasmalemmal vesicles, cytoplasmic expansion, and capping. Degenerative changes included enlargement of mitochondria and effacement of cristae, and disintegration of both abaxonal and adaxonal cytosol and organelles. Both demyelination and onion-bulb formation were seen at all time points, although supernumerary Schwann cells and axonal degeneration were most numerous after 24 months of galactose feeding. In sural nerve biopsy samples from patients with diabetes and progressive worsening of neuropathy, ultrastructural abnormalities in Schwann cells encompassed the full range of reactive, degenerative and proliferative changes described in galactose-fed rats. The concordance of fine-structural observations in nerves from galactose-fed rats and these adult-onset diabetic patients emphasizes the role of flux through aldose reductase in the complex pathology of diabetic neuropathy and points to the utility of galactose intoxication in helping to understand this metabolic disorder.


Subject(s)
Diabetic Neuropathies/pathology , Galactose/toxicity , Peripheral Nervous System Diseases/pathology , Schwann Cells/physiology , Adult , Aged , Animals , Cell Division/physiology , Edema/pathology , Female , Humans , Male , Mast Cells/drug effects , Mast Cells/ultrastructure , Microscopy, Electron , Middle Aged , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Peripheral Nervous System Diseases/chemically induced , Rats , Rats, Sprague-Dawley , Schwann Cells/drug effects , Sural Nerve/pathology
18.
Eur J Immunol ; 27(11): 2863-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9394811

ABSTRACT

Previous studies examining the effect of nitric oxide synthase (NOS) inhibition on the course of experimental allergic encephalomyelitis (EAE) have yielded conflicting results. This may relate to the use of nonspecific inhibitors and to differences between active and adoptive EAE. We examined the effect of treatment with L-N-(1-iminoethyl)lysine (L-NIL), a selective inhibitor of the cytokine-inducible isoform of NOS, on the clinical course of active and adoptive EAE in Lewis rats. We find that while L-NIL treatment of recipients is protective in adoptive EAE, treatment of active EAE with L-NIL leads to a marked accentuation of disease expression. In L-NIL-treated animals treated with myelin basic protein/complete Freund's adjuvant (MBP/CFA), disease onset is accelerated and clinical symptoms are more severe. Accentuation of integrated disease scores is seen even if L-NIL treatment is started 5 days following immunization. The histological findings in involved spinal cords from L-NIL-treated animals with active EAE are similar to those from untreated animals with similar clinical scores. L-NIL treatment of MBP/CFA-immunized animals does not prevent recovery from clinical symptoms, nor does it allow for reinduction of disease in animals previously immunized with MBP/CFA. Treatment of F344 rats, a strain which is relatively nonsusceptible for EAE, with L-NIL results in consistent evidence of EAE following immunization with MBP/CFA. These findings, together with our previous work on interstitial nephritis, support a role for endogenously generated NO in immunoregulation of T cell responses following immunization with antigen in CFA, and suggest that inducibility of NOS expression may be an important susceptibility factor for autoimmunity.


Subject(s)
Adjuvants, Immunologic/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Nitric Oxide/immunology , Nitric Oxide/physiology , Adjuvants, Immunologic/biosynthesis , Administration, Oral , Adoptive Transfer , Animals , Concanavalin A/pharmacology , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Freund's Adjuvant/immunology , Immunity, Innate/drug effects , Interferon-gamma/biosynthesis , Interferon-gamma/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Transfusion , Lysine/administration & dosage , Lysine/analogs & derivatives , Myelin Basic Protein/immunology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Species Specificity , Spleen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
19.
Neurosci Lett ; 234(1): 15-8, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9347935

ABSTRACT

Transgenic mice expressing interleukin-6 (IL6) in the brain exhibit gliosis, spongiosis and neuronal loss. Based on previous findings, we hypothesized that IL6 could upregulate the prion protein (PrP) gene in the central nervous system (CNS) of these mice. Western and Northern blot analysis showed that PrP protein and mRNA levels were comparable to control levels. Furthermore, ultrastructural characterization revealed that spongiosis was actually located in astrocytes. These results indicate that IL6 does not upregulate the cerebral PrP expression in this animal model and that profound astrocytic alterations precipitate the neuronal degeneration observed.


Subject(s)
Brain/metabolism , Interleukin-6/biosynthesis , Prion Diseases/physiopathology , Prions/physiology , Animals , Glial Fibrillary Acidic Protein/analysis , Immunoblotting , Mice , Mice, Transgenic , Microscopy, Electron , Prion Diseases/etiology
20.
Acta Neuropathol ; 93(6): 611-8, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9194901

ABSTRACT

In galactose neuropathy, aldose reductase inhibitor (ARI)-preventable Schwann cell injury has been reported in studies in which galactose feeding continued over a period of months. Given the link between these morphologic changes and polyol pathway flux, polyol accumulation after just days of galactose feeding points to the possibility that structural changes occur much earlier than previously reported. The aim of this study was to examine rat sciatic nerve after 7 days of galactose feeding for evidence of myelinated fiber injury and establish whether it is related to polyol accumulation. Compared to control or ARI-treated galactose-fed rats, nerves from untreated galactose-fed rats had increased water (P < 0.05) ad dulcitol (P < 0.008) content and decreased amounts of myo-inositol (P < 0.01). Electron microscopy revealed reactive Schwann cell changes in myelinated fibers characterized by increased cytoplasmic volume, and the occurrence of lipid droplets pi granules of Reich and enlarged mitochondria. Dystrophic accumulation of intermediate filaments was also observed in the inner glial loop. Degenerative changes included periaxonal swelling, enlarged mitochondria without recognizable cristae, lysis of Schwann cell cytoplasm and demyelination. Reactive (P < 0.05) and degenerative (P < 0.01) changes as well as the number of redundant basal lamina profiles (P < 0.05) were significantly more frequent in untreated galactose-fed rats compared to controls. ARI treatment attenuated these changes. Consistent with the initial stages of onion-bulb formation, profiles with imbricate Schwann cells were also seen only in untreated galactose-fed rats. The findings suggest that short-term increases in polyol pathway activity can have deleterious effects Schwann cells of myelinated fibers.


Subject(s)
Galactose/toxicity , Schwann Cells/drug effects , Schwann Cells/pathology , Administration, Oral , Aldehyde Reductase/antagonists & inhibitors , Animals , Diabetic Neuropathies/chemically induced , Diabetic Neuropathies/pathology , Drug Administration Schedule , Female , Rats , Rats, Sprague-Dawley , Schwann Cells/ultrastructure , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...