Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Neuroscience ; 319: 206-20, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26777891

ABSTRACT

Exposure to blast overpressure (BOP) is associated with behavioral, cognitive, and neuroimaging abnormalities. We investigated the dynamic responses of cortical vasculature and its relation to microglia/macrophage activation in mice using intravital two-photon microscopy following mild blast exposure. We found that blast caused vascular dysfunction evidenced by microdomains of aberrant vascular permeability. Microglial/macrophage activation was specifically associated with these restricted microdomains, as evidenced by rapid microglial process retraction, increased ameboid morphology, and escape of blood-borne Q-dot tracers that were internalized in microglial/macrophage cell bodies and phagosome-like compartments. Microdomains of cortical vascular disruption and microglial/macrophage activation were also associated with aberrant tight junction morphology that was more prominent after repetitive (3×) blast exposure. Repetitive, but not single, BOPs also caused TNFα elevation two weeks post-blast. In addition, following a single BOP we found that aberrantly phosphorylated tau rapidly accumulated in perivascular domains, but cleared within four hours, suggesting it was removed from the perivascular area, degraded, and/or dephosphorylated. Taken together these findings argue that mild blast exposure causes an evolving CNS insult that is initiated by discrete disturbances of vascular function, thereby setting the stage for more protracted and more widespread neuroinflammatory responses.


Subject(s)
Blast Injuries/pathology , Brain Injuries/pathology , Macrophages/pathology , Microglia/pathology , Animals , Blood-Brain Barrier/pathology , Blotting, Western , Brain/blood supply , Brain/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Immunohistochemistry , Intravital Microscopy , Male , Mice , Mice, Inbred C57BL , Microvessels/pathology
2.
J Microsc ; 241(2): 153-61, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21118209

ABSTRACT

A benefit of multiphoton fluorescence microscopy is the inherent optical sectioning that occurs during excitation at the diffraction-limited spot. The scanned collection of fluorescence emission is incoherent; that is, no real image needs to be formed on the detector plane. The nearly isotropic emission of fluorescence excited at the focal spot allows for new detection schemes that efficiently funnel all attainable photons to detector(s). We previously showed [Combs, C.A., et al. (2007) Optimization of multiphoton excitation microscopy by total emission detection using a parabolic light reflector. J. Microsc. 228, 330-337] that parabolic mirrors and condensers could be combined to collect the totality of solid angle around the excitation spot for tissue blocks, leading to ∼8-fold signal gain. Using a similar approach, we have developed an in vivo total emission detection (epiTED) instrument modified to make noncontact images from outside of living tissue. Simulations suggest that a ∼4-fold enhancement may be possible (much larger with lower NA objectives than the 0.95 NA used here) with this approach, depending on objective characteristics, imaging depth and the characteristics of the sample being imaged. In our initial prototype, 2-fold improvements were demonstrated in the mouse brain and skeletal muscle as well as the rat kidney, using a variety of fluorophores and no compromise of spatial resolution. These results show this epiTED prototype effectively doubles emission signal in vivo; thus, it will maintain the image signal-to-noise ratio at two times the scan rate or enable full scan rate at approximately 30% reduced laser power (to minimize photo-damage).


Subject(s)
Microscopy, Fluorescence, Multiphoton/methods , Animals , Brain/cytology , Brain Chemistry , Image Processing, Computer-Assisted/methods , Kidney/chemistry , Kidney/cytology , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/chemistry , Muscle, Skeletal/cytology , Rats , Rats, Wistar
3.
J Cell Biol ; 155(2): 301-10, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11604425

ABSTRACT

alpha-Dystroglycan (DG) has been identified as the cellular receptor for lymphocytic choriomeningitis virus (LCMV) and Lassa fever virus (LFV). This subunit of DG is a highly versatile cell surface molecule that provides a molecular link between the extracellular matrix (ECM) and a beta-DG transmembrane component, which interacts with the actin-based cytoskeleton. In addition, DG exhibits a complex pattern of interaction with a wide variety of ECM and cellular proteins. In the present study, we characterized the binding of LCMV to alpha-DG and addressed the role of alpha-DG-associated host-derived proteins in virus infection. We found that the COOH-terminal region of alpha-DG's first globular domain and the NH2-terminal region of the mucin-related structures of alpha-DG together form the binding site for LCMV. The virus-alpha-DG binding unlike ECM alpha-DG interactions was not dependent on divalent cations. Despite such differences in binding, LCMV and laminin-1 use, in part, an overlapping binding site on alpha-DG, and the ability of an LCMV isolate to compete with laminin-1 for receptor binding is determined by its binding affinity to alpha-DG. This competition of the virus with ECM molecules for receptor binding likely explains the recently found correlation between the affinity of LCMV binding to alpha-DG, tissue tropism, and pathological potential. LCMV strains and variants with high binding affinity to alpha-DG but not low affinity binders are able to infect CD11c+ dendritic cells, which express alpha-DG at their surface. Infection followed by dysfunction of these antigen-presenting cells contributes to immunosuppression and persistent viral infection in vivo.


Subject(s)
Cytoskeletal Proteins/metabolism , Lymphocytic choriomeningitis virus/metabolism , Lymphocytic choriomeningitis virus/pathogenicity , Membrane Glycoproteins/metabolism , Animals , Arenaviridae Infections/metabolism , Arenaviridae Infections/virology , Binding Sites , Binding, Competitive , Cells, Cultured , Cytoskeletal Proteins/chemistry , Dystroglycans , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Female , Laminin/metabolism , Lymphocytic choriomeningitis virus/isolation & purification , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Spleen/metabolism , Spleen/virology
4.
Brain ; 124(Pt 7): 1403-16, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408335

ABSTRACT

A major question in neurobiology is whether myelin repair can restore neurological function following the course of a severe, progressive CNS demyelinating disease that induces axonal loss. In this study we used Theiler's murine encephalomyelitis virus (TMEV) to induce a chronic progressive CNS demyelinating disease in mice that was immune-mediated and pathologically similar to human multiple sclerosis. Because immunosuppression of chronically TMEV-infected mice has been shown to enhance myelin repair, we first addressed the potential roles of CD4(+) and CD8(+) T cells in the inhibition of CNS remyelination during chronic disease. TMEV infection of susceptible PL/J mice deficient in CD4(+) but not CD8(+) T cells demonstrated a significant increase in severity of pathogenesis when compared with wild-type controls. This was characterized by enhanced demyelination, spinal cord atrophy, neurological deficits, and mortality. Interestingly, the PL/J CD4(-/-) mice that survived to the chronic stage of the disease had nearly complete spontaneous myelin repair mediated by both oligodendrocytes and infiltrating Schwann cells. Therefore, we next addressed whether this spontaneous myelin repair was associated with improved neurological function despite the increased pathology. Of interest, all surviving PL/J CD4(-/-) mice showed partial restoration of motor coordination and gait that coincided temporally with spontaneous myelin repair. Furthermore, functional recovery of motor coordination correlated strongly with the percentage of myelin repair mediated by Schwann cells, whereas restoration of hindlimb gait correlated with oligodendrocyte-mediated myelin repair. This is the first study to demonstrate that spontaneous remyelination correlates with partial restoration of neurological function during the course of a progressive, immune-mediated CNS demyelinating disease. Of greater importance, functional recovery occurred despite previous severe demyelination and spinal cord atrophy.


Subject(s)
Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Myelin Sheath/metabolism , Recovery of Function , Animals , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , Cardiovirus Infections/immunology , Cardiovirus Infections/pathology , Cardiovirus Infections/physiopathology , Cardiovirus Infections/virology , Chronic Disease , Disease Models, Animal , Disease Progression , Mice , Mice, Knockout , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Remission, Spontaneous , Severity of Illness Index , Spinal Cord/pathology , Survival Rate , Theilovirus/pathogenicity
5.
Am J Pathol ; 157(4): 1365-76, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11021839

ABSTRACT

Infection of susceptible mice with Theiler's murine encephalomyelitis virus results in neurological dysfunction from progressive central nervous system demyelination that is pathologically similar to the human disease, multiple sclerosis. We hypothesized that the development of neuropathology proceeds down a final common pathway that can be accurately quantified within a single spinal cord lesion. To test this hypothesis, we conducted quantitative ultrastructural analyses of individual demyelinated spinal cord lesions from chronically infected mice to determine whether pathological variables assessed within a single lesion accurately predicted global assessments of morphological and functional disease course. Within lesions we assessed by electron microscopy the frequencies of normally myelinated, remyelinated, and demyelinated axons, as well as degenerating axons and intra-axonal mitochondria. The frequency of medium and large remyelinated fibers within a single lesion served as a powerful indicator of axonal preservation and correlated with preserved neurological function. The number of degenerating axons and increased intra-axonal mitochondria also correlated strongly with global measures of disease course, such as total lesion load, spinal cord atrophy, and neurological function. This is the first study to demonstrate that functional severity of disease course is evident within a single demyelinated lesion analyzed morphometrically at the ultrastructural level.


Subject(s)
Demyelinating Diseases/pathology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Nervous System/physiopathology , Spinal Cord Diseases/pathology , Spinal Cord/ultrastructure , Animals , Atrophy , Axons/ultrastructure , Cardiovirus Infections/pathology , Cardiovirus Infections/physiopathology , Demyelinating Diseases/physiopathology , Disease Models, Animal , Forecasting , Mice , Mice, Inbred Strains , Mitochondria/ultrastructure , Nerve Degeneration/pathology , Spinal Cord Diseases/physiopathology , Theilovirus
6.
Brain Res ; 877(2): 396-400, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10986359

ABSTRACT

Identifying the role of axonal injury in the development of permanent, irreversible neurologic disability is important to the study of central nervous system (CNS) demyelinating diseases. Our understanding of neurologic dysfunction in demyelinating diseases and the ability to assess therapeutic interventions depends on the development of objective functional assays that can non-invasively measure axonal loss. In this study, we demonstrate in a murine model of progressive CNS demyelination that assessment of the hindlimb width of stride provides a powerful indicator of axonal loss and can dissociate between deficits induced by demyelination versus axonal loss.


Subject(s)
Axons/pathology , Central Nervous System/pathology , Demyelinating Diseases/pathology , Lameness, Animal/pathology , Nerve Degeneration/pathology , Nerve Fibers, Myelinated/pathology , Animals , Axons/ultrastructure , Axons/virology , Biomarkers , Cardiovirus Infections/pathology , Cardiovirus Infections/physiopathology , Central Nervous System/ultrastructure , Central Nervous System/virology , Demyelinating Diseases/physiopathology , Demyelinating Diseases/virology , Disease Models, Animal , Female , Lameness, Animal/physiopathology , Lameness, Animal/virology , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/physiopathology , Nerve Degeneration/virology , Nerve Fibers, Myelinated/ultrastructure , Nerve Fibers, Myelinated/virology , Theilovirus/physiology
7.
Mol Cell Neurosci ; 15(6): 495-509, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10860577

ABSTRACT

The importance of genetic susceptibility in determining the progression of demyelination and neurologic deficits is a major focus in neuroscience. We studied the influence of human leukocyte antigen (HLA)-DQ polymorphisms on disease course and neurologic impairment in virus-induced demyelination. HLA-DQ6 or DQ8 was inserted as a transgene into mice lacking endogenous expression of MHC class I (beta(2)m) and class II (H2-A(beta)) molecules. Following Theiler's murine encephalomyelitis virus (TMEV) infection, we assessed survival, virus persistence, demyelination, and clinical disease. Mice lacking expression of endogenous class I and class II molecules (beta(2)m(o) Abeta(o) mice) died 3 to 4 weeks postinfection (p.i.) due to overwhelming virus replication in neurons. beta(2)m(o) Abeta(o) DQ6 and beta(2)m(o) Abeta(o) DQ8 mice had increased survival and decreased gray matter disease and virus replication compared to nontransgenic littermate controls. Both beta(2)m(o) Abeta(o) DQ6 and beta(2)m(o) Abeta(o) DQ8 mice developed chronic virus persistence in glial cells of the white matter of the spinal cord, with greater numbers of virus antigen-positive cells in beta(2)m(o) Abeta(o) DQ8 than in beta(2)m(o) Abeta(o) DQ6 mice. At day 45 p.i., the demyelinating lesions in the spinal cord of beta(2)m(o) Abeta(o) DQ8 were larger than those in the beta(2)m(o) Abeta(o) DQ6 mice. Earlier and more profound neurologic deficits were observed in beta(2)m(o) Abeta (o) DQ8 mice compared to beta(2)m(o) Abeta(o) DQ6 mice, although by 120 days p.i. both strains of mice showed similar extent of demyelination and neurologic deficits. Delayed-type hypersensitivity and antibody responses to TMEV demonstrated that the mice mounted class II-mediated cellular and humoral immune responses. The results are consistent with the hypothesis that rates of progression of demyelination and neurologic deficits are related to the differential ability of DQ6 and DQ8 transgenes to modulate the immune response and control virus.


Subject(s)
Cardiovirus Infections/genetics , HLA-DQ Antigens/genetics , Multiple Sclerosis/genetics , Polymorphism, Genetic , Theilovirus , Acute Disease , Animals , Antibody Formation/genetics , Antigens, Viral/analysis , Brain/immunology , Brain/virology , Cardiovirus Infections/immunology , Cardiovirus Infections/mortality , Chronic Disease , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Humans , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity , Multiple Sclerosis/immunology , Nerve Fibers/immunology , Nerve Fibers/virology , Postural Balance , Spinal Cord/immunology , Spinal Cord/virology , Survival Analysis , Virus Replication/immunology
8.
Brain ; 123 Pt 3: 519-31, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10686175

ABSTRACT

Recent pathological studies have re-emphasized that axonal injury is present in patients with multiple sclerosis, the most common demyelinating disease of the CNS in humans. However, the temporal profile of demyelination and axonal loss in multiple sclerosis patients and their independent contributions to clinical and electrophysiological abnormalities are not completely understood. In this study, we used the Theiler's murine encephalomyelitis virus model of progressive CNS inflammatory demyelination to demonstrate that demyelination in the spinal cord is followed by a loss of medium to large myelinated fibres. By measuring spinal cord areas, motor-evoked potentials, and motor coordination and balance, we determined that axonal loss following demyelination was associated with electrophysiological abnormalities and correlated strongly with reduced motor coordination and spinal cord atrophy. These findings demonstrate that axonal loss can follow primary, immune-mediated demyelination in the CNS and that the severity of axonal loss correlates almost perfectly with the degree of spinal cord atrophy and neurological deficits.


Subject(s)
Axons/pathology , Multiple Sclerosis/pathology , Poliomyelitis/pathology , Spinal Cord Diseases/pathology , Theilovirus , Animals , Atrophy , Disease Models, Animal , Evoked Potentials, Motor , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Nerve Fibers, Myelinated/immunology , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/virology , Neural Conduction , Neurologic Examination , Poliomyelitis/immunology , Psychomotor Performance , Spinal Cord/pathology , Spinal Cord Diseases/immunology , Spinal Cord Diseases/virology
9.
J Neurosci Res ; 58(4): 492-504, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10533042

ABSTRACT

Spinal cord pathology, such as demyelination and axonal loss, is a common feature in multiple models of central nervous system (CNS) injury and disease. Development of methods to quantify spinal cord pathology objectively would aid studies designed to establish mechanisms of damage, correlate pathology with neurologic function, and assess therapeutic interventions. In this study, we describe sensitive methods to objectively quantify spinal cord demyelination, remyelination, atrophy, and axonal loss following the initiation of a progressive inflammatory demyelinating disease with Theiler's murine encephalomyelitis virus (TMEV). Spinal cord demyelination, remyelination, and atrophy were quantified from representative 1-microm-thick cross sections embedded in Araldite plastic using interactive image analysis. In addition, this study demonstrates novel, automated methodology to quantify axonal loss from areas of normal-appearing white matter, as a measure of secondary axonal injury following demyelination. These morphologic methods, which are applicable to various models of CNS injury, provide an innovative way to assess the benefits of therapeutic agents, to determine mechanisms of spinal cord damage, or to establish a correlation with sensitive measures of neurologic function. J. Neurosci Res 58:492-504.


Subject(s)
Axons/pathology , Demyelinating Diseases/pathology , Myelin Sheath/pathology , Neurons/pathology , Poliomyelitis/pathology , Spinal Cord/pathology , Theilovirus , Animals , Atrophy , Cell Count , Central Nervous System/pathology , Mice , Mice, Inbred Strains , Mice, Knockout , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
10.
Exp Neurol ; 158(1): 171-81, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10448429

ABSTRACT

The precise factors involved in the development of a progressive motor dysfunction, a hallmark of immune-mediated demyelinating diseases such as multiple sclerosis, are not well defined. The ability to identify neurologic deficits that result in impaired motor performance early in disease may allow for the identification of therapeutic interventions that slow or eliminate the progression toward a permanent dysfunction. Here we describe the use of three objective, quantitative functional assays (spontaneous activity box, rotarod, and footprint analysis) to detect early neurologic deficits following the initiation of a demyelinating disease with Theiler's murine encephalomyelitis virus (TMEV). The results show that the assays are capable of revealing neurologic deficits at the early stages of the demyelinating disease process. These findings are the first to objectively characterize neurologic function in an animal model of progressive CNS demyelination.


Subject(s)
Central Nervous System/pathology , Disease Models, Animal , Movement Disorders/diagnosis , Movement Disorders/etiology , Multiple Sclerosis/complications , Multiple Sclerosis/pathology , Acute Disease , Analysis of Variance , Animals , Cardiovirus Infections/complications , Central Nervous System/virology , Chronic Disease , Disease Progression , Methods , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Multiple Sclerosis/virology , Theilovirus/physiology , Time Factors
11.
J Neurosci ; 18(18): 7306-14, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9736651

ABSTRACT

In this study we demonstrate perforin-mediated cytotoxic effector function is necessary for viral clearance and may directly contribute to the development of neurologic deficits after demyelination in the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis. We previously demonstrated major histocompatability complex (MHC) class I-deficient (beta2m-deficient) mice with an otherwise resistant genotype develop severe demyelination with minimal neurologic disease when chronically infected with TMEV. These studies implicate CD8(+) T cells as the pathogenic cell in the induction of neurologic disease after demyelination. To determine which effector mechanisms of CD8(+) T cells, granule exocytosis or Fas ligand expression, play a role in the development of demyelination and clinical disease, we infected perforin-deficient, lpr (Fas mutation), and gld (Fas ligand mutation) mice with TMEV. Perforin-deficient mice showed viral persistence in the CNS, chronic brain pathology, and demyelination in the spinal cord white matter. Perforin-deficient mice demonstrated severely impaired MHC class I-restricted cytotoxicity against viral epitopes, but normal MHC class II-restricted delayed-type hypersensitivity responses to virus antigen. Despite demyelination, virus-infected perforin-deficient mice showed only minimal neurologic deficits as indicated by clinical disease score, activity monitoring, and footprint analysis. Perforin- and MHC class II-deficient mice (with functional CD8(+) T cells and perforin molecules and an H-2(b) haplotype) had comparable demyelination and genotype, however, only the latter showed severe clinical disease. Gld and lpr mice demonstrated normal TMEV-specific cytotoxicity and maintained resistance to TMEV-induced demyelinating disease. These studies implicate perforin release by CD8(+) T cells as a potential mechanism by which neurologic deficits are induced after demyelination.


Subject(s)
Membrane Glycoproteins/immunology , Multiple Sclerosis/physiopathology , Neurons/virology , Poliomyelitis/physiopathology , Theilovirus , Animals , Chronic Disease , Demyelinating Diseases/immunology , Demyelinating Diseases/virology , Disease Models, Animal , Exocytosis/immunology , Fas Ligand Protein , Female , Histocompatibility Antigens Class I/immunology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron , Multiple Sclerosis/immunology , Mutation , Neurons/chemistry , Neurons/physiology , Perforin , Poliomyelitis/immunology , Pore Forming Cytotoxic Proteins , Spinal Cord/cytology , Spinal Cord/ultrastructure , T-Lymphocytes, Cytotoxic/chemistry , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...