Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
J Virol ; 74(21): 10217-22, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11024152

ABSTRACT

Using a yeast two-hybrid human brain cDNA library screen, the cytoplasmic dynein light chain (LC8), a 10-kDa protein, was found to interact strongly with the phosphoprotein (P) of two lyssaviruses: rabies virus (genotype 1) and Mokola virus (genotype 3). The high degree of sequence divergence between these P proteins (only 46% amino acid identity) favors the hypothesis that this interaction is a common property shared by all lyssaviruses. The P protein-dynein LC8 interaction was confirmed by colocalization with laser confocal microscopy in infected cells and by coimmunoprecipitation. The dynein-interacting P protein domain was mapped to the 186 amino acid residues of the N-terminal half of the protein. Dynein LC8 is a component of both cytoplasmic dynein and myosin V, which are involved in a wide range of intracellular motile events, such as microtubule minus-end directed organelle transport in axon "retrograde transport" and actin-based vesicle transport, respectively. Our results provide support for a model of viral nucleocapsid axoplasmic transport. Furthermore, the role of LC8 in cellular mechanisms other than transport, e.g., inhibition of neuronal nitric oxide synthase, suggests that the P protein interactions could be involved in physiopathological mechanisms of rabies virus-induced pathogenesis.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins , Lyssavirus/metabolism , Phosphoproteins/metabolism , Viral Proteins/metabolism , Animals , Brain/metabolism , Cell Line , Cytoplasm/physiology , DNA, Complementary , Dyneins , Gene Library , Humans , Lyssavirus/genetics , Microscopy, Confocal , Molecular Chaperones , Phosphoproteins/genetics , Precipitin Tests , Rabies virus/genetics , Rabies virus/metabolism , Rhabdoviridae Infections/virology , Two-Hybrid System Techniques , Viral Proteins/genetics , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism
2.
Rev Neurol (Paris) ; 156(3): 236-41, 2000 Mar.
Article in French | MEDLINE | ID: mdl-10740094

ABSTRACT

Rabies virus variants obtained by recombinant DNA techniques enabled us to use the high neurotropism of rabies virus to express foreign genes (e.g: Chloramphenicol Acetyl Transferase gene) in neuronal cell cultures as well as in rodent brain. The foreign gene was inserted in the viral pseudogene region; this insertion did not affect the neurotropism of rabies virus, as shown by infection of neuronal cell cultures without any major cytopathic effects for several days. Stereotaxic inoculation of these rabies virus variants into rat striatum indicated that insertion of the foreign gene did not alter the viral axonal transport and the subsequent widespread brain infection. These data allow to consider rabies virus as a vector for the selective expression of foreign genes in neurons.


Subject(s)
Brain/virology , Neurons/immunology , Rabies virus/genetics , Vaccines, DNA/genetics , Animals , Brain/immunology , Brain/pathology , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , DNA Transposable Elements/genetics , Gene Expression , Genes, Viral/genetics , Genes, Viral/immunology , Genetic Variation , Genetic Vectors/genetics , In Vitro Techniques , Mice , Neurons/virology , Pseudogenes/genetics , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies virus/immunology , Rats , Transcription, Genetic/genetics , Tropism/genetics , Vaccines, DNA/immunology
3.
J Neurovirol ; 4(1): 115-9, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9531019

ABSTRACT

We investigated the infection characteristics of recombinant rabies virus variants modified in the pseudogene sequence. Infection of neuronal cell lines by the SAD W9 and SAD V* variants (respectively with deletion or insertion in this sequence) showed no significant differences as compared to the parental strain, the attenuated strain SAD B19, in infection characteristics such as number of infected cells or viral yield. The inoculation of mice by these variants resulted in similar infection patterns and pathogenicity. Stereotaxic inoculation of the different variants into the rat striatum showed that deletion or insertion did not affect the axonal virus spread, nor did insertion of a complete additional transcription unit, that could be expressed in the areas connected to the inoculation site. These results show that the pseudogene sequence is not involved in viral spread and pathogenicity and confirm the availability of this domain for targeting and expression of foreign genes into neurons.


Subject(s)
Genes, Viral/genetics , Pseudogenes/genetics , Rabies virus/genetics , Animals , Axonal Transport/physiology , Cells, Cultured , DNA Transposable Elements , Mice , Models, Biological , Neurons/virology , Rabies virus/growth & development , Rats , Sequence Deletion , Stereotaxic Techniques
4.
J Virol ; 72(1): 823-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9420294

ABSTRACT

Apoptosis has been suggested as a mechanism by which dengue (DEN) virus infection may cause neuronal cell death (P. Desprès, M. Flamand, P.-E. Ceccaldi, and V. Deubel, J. Virol. 70:4090-4096, 1996). In this study, we investigated whether apoptotic cell death occurred in the central nervous system (CNS) of neonatal mice inoculated intracerebrally with DEN virus. We showed that serial passage of a wild-type human isolate of DEN virus in mouse brains selected highly neurovirulent variants which replicated more efficiently in the CNS. Infection of newborn mice with these neurovirulent variants produced fatal encephalitis within 10 days after inoculation. Virus-induced cell death and oligonucleosomal DNA fragmentation were observed in mouse brain tissue by day 9. Infected mouse brain tissue was assayed for apoptosis by in situ terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and for virus replication by immunostaining of viral antigens and in situ hybridization. Apoptotic cell death and DEN virus replication were restricted to the neurons of the cortical and hippocampal regions. Thus, DEN virus-induced apoptosis in the CNS was a direct result of virus infection. In the murine neuronal cell line Neuro 2a, neuroadapted DEN virus variants showed infection patterns similar to those of the parental strain. However, DEN virus-induced apoptosis in these cells was more pronounced after infection with the neurovirulent variants than after infection with the parental strain.


Subject(s)
Apoptosis , Brain/pathology , Brain/virology , Dengue Virus/pathogenicity , Dengue/pathology , Dengue/virology , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Animals , Animals, Newborn , Cerebral Cortex/pathology , Cerebral Cortex/virology , DNA Fragmentation , Dengue Virus/genetics , Dengue Virus/physiology , Hippocampus/pathology , Hippocampus/virology , Humans , In Situ Hybridization , Mice , RNA, Viral/genetics , RNA, Viral/metabolism , Virulence , Virus Replication
5.
J Gen Virol ; 78 ( Pt 11): 2831-5, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9367369

ABSTRACT

We investigated the effect of rabies virus infection on the actin cytoskeleton using various techniques. Confocal microscopic examination of rabies virus-infected neuroblastoma cells at late stages of infection revealed a dramatic decrease in F-actin staining. The results of a fluorimetric assay with pyrenylated actin indicated that purified rabies virus nucleocapsid has no direct action on the kinetics of actin polymerization and only a weak effect on the final extent of polymerization. Video-microscopy experiments with purified components showed that rabies virus nucleocapsid inhibits the actin-bundling effect induced by dephospho-synapsin I, a neuron-specific protein which is known to exert a control on the actin-based cytoskeleton. Thus, the observed decrease in F-actin staining in infected cells might be ascribed to an indirect action of rabies nucleocapsid on the effects of actin-binding proteins such as synapsin I.


Subject(s)
Actins/metabolism , Cytoskeleton/pathology , Rabies virus , Rabies/pathology , Dimerization , Humans , Microscopy, Confocal , Rabies/metabolism , Tumor Cells, Cultured
6.
FEBS Lett ; 398(2-3): 211-6, 1996 Dec 02.
Article in English | MEDLINE | ID: mdl-8977109

ABSTRACT

We have analyzed the effects of synaptic vesicles on actin polymerization by using a time-resolved spectrofluorometric assay. We have found that synaptic vesicles have complex effects on the kinetics of actin polymerization, which vary depending on whether the synaptic vesicle-specific phosphoprotein synapsin I is absent or present on their membrane. Synapsin I bound either to synaptic vesicles or to pure phospholipid vesicles exhibits phosphorylation-dependent actin-nucleating activity. Synaptic vesicles depleted of endogenous synapsin I decrease the rate and the final extent of actin polymerization, an effect which is not observed with pure phospholipid vesicles. Thus, the state of association of synapsin I with synaptic vesicles, which is modulated by its state of phosphorylation, may affect actin assembly and the physico-chemical characteristics of the synaptic vesicle microenvironment.


Subject(s)
Actins/metabolism , Synapsins/metabolism , Synaptic Vesicles/metabolism , Animals , Cattle , Kinetics , Membranes, Artificial , Osmolar Concentration , Phosphorylation , Polymers , Rats , Spectrometry, Fluorescence , Synaptic Vesicles/chemistry , Viscosity
7.
J Neuroimmunol ; 68(1-2): 45-51, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8784259

ABSTRACT

Interleukin-1 beta (IL-1 beta) and tumor necrosis factor-alpha (TNF alpha) are important cytokines in the development of brain inflammation during pathological process. During rabies virus infection, the level of these proinflammatory cytokines are enhanced in the brain. In the present study we determined the cellular localization of these two cytokines by immunocytochemistry in brains of rats infected with rabies virus, at different time-intervals of the disease (day 1, 3, 4, 5 and at final stage day 6 post-infection (p.i.)). Cellular identification of IL-1 beta (irIL-1 beta) and TNF alpha (irTNF alpha) immunopositive cells was studied using a polyclonal antibody against these cytokines and against glial fibrillary acidic protein (GFAP) to detect astrocytes and GSA-I-B4 isolectin to detect microglial cells and/or infiltrating macrophages. In brains of control and early infected rats, irIL-1 beta was only detected in fibers located in the hypothalamus, supraoptic and tractus optic nuclei and infundibular nucleus. From day 4 onwards until day 6 p.i., enhanced irIL-1 beta was found and identified either in activated ameboid and/or infiltrated macrophages (amygdala, thalamus, internal capsula, subtantia nigra, septal nuclei and around blood vessels), or in activated ramified cells (hypothalamus and periventricular nucleus, piriformis and cingulate cortex, hippocampus). IrTNF alpha was observed in the brains of rats at a final stage of disease (day 5 and 6 p.i.): in the hypothalamus, the amygdala, the internal capsula, the thalamus, the septal nuclei, the hippocampus, the habenular nuclei and around the blood vessels. Ir-TNF alpha was detected in round cells identified as ameboid microglia and/or infiltrated macrophages. A marked activation of microglial and astroglial cells was observed mainly in the hypothalamus, the thalamus and hippocampus and around the blood vessels, at day 4 p.i. and later, revealing a high central inflammatory reaction in brains of rabies virus infected rats. These results showed that IL-1 beta and TNF alpha are produced in the brain both by local microglial cells and infiltrating macrophages during rabies infection. Thus, these cytokines may play an important role in coordinating the dramatic inflammatory response associated with the rabies-encephalopathy as well as in the neural modification and alteration of brain functions.


Subject(s)
Brain/immunology , Interleukin-1/immunology , Rabies virus/immunology , Rabies/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Animals, Suckling , Brain/virology , Brain Chemistry/immunology , Male , Mice , Microglia/chemistry , Microglia/immunology , Microglia/virology , Rabbits , Rats , Rats, Wistar
8.
Int J Radiat Biol ; 70(1): 69-75, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8691037

ABSTRACT

Ionizing radiation has been shown to affect a broad range of viral diseases including neurotropic infections through an immunosuppression mechanism. In the present study we have investigated the effect of ionizing radiation on the characteristics of neurotropic infection by rabies virus, which has the unusual feature of infecting almost exclusively neurons. In order to analyze better the effect produced, the study concerned the spread of an apathogenic rabies virus variant in mouse brain. Irradiation was shown to increase both the intensity and duration of the infection in a reversible and dose-dependent manner and was effective in whole-body irradiation and in head-protected body irradiation, whereas cephalic irradiation had no effect. These results underline the role played by the immune system in the regulation of neurotropic virus infections in the brain and show that phenomena such as viral clearance and time-course of a neurotropic viral infection may be significantly modified by ionizing radiation, even for viruses whose infection involves only neurons.


Subject(s)
Brain/virology , Immune System/radiation effects , Rabies/immunology , Animals , Brain/radiation effects , Dose-Response Relationship, Radiation , Male , Mice , Mice, Inbred BALB C , Radiation Protection
9.
J Virol ; 70(6): 4090-6, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8648748

ABSTRACT

Human isolates of dengue (DEN) type 1 viruses FGA/89 and BR/90 differ in their membrane fusion properties in mosquito cell lines (P. Desprès et al., Virology 196:209-216, 1993). FGA/89 and BR/90 were assayed for their neurovirulence in newborn mice, and neurons were the major target cells for both DEN-1 virus strains within the central nervous system. To study the susceptibility of neurons to DEN virus infection, DEN virus replication was analyzed in the murine neuroblastoma cell line Neuro 2a. Infection of Neuro 2a cells with FGA/89 or BR/90 induced apoptotic DNA degradation after 25 h of infection. Studies of DEN protein synthesis revealed that accumulation of viral proteins leads to apoptotic cell death. The apoptotic process progressed more rapidly following BR/90 infection than it did after FGA/89 infection. The higher cytotoxicity of BR/90 for Neuro 2a cells was linked to an incomplete maturation of the envelope proteins, resulting in abortive virus assembly. Accumulation of viral proteins in the endoplasmic reticulum may induce stress and thereby activate the apoptotic pathway in mouse neuroblastoma cells.


Subject(s)
Apoptosis , Dengue Virus/physiology , Neuroblastoma/pathology , Animals , Chlorocebus aethiops , DNA/metabolism , Humans , Mice , NF-kappa B/metabolism , Vero Cells , Virus Assembly
10.
Arch Virol ; 141(3-4): 573-85, 1996.
Article in English | MEDLINE | ID: mdl-8645096

ABSTRACT

We have evaluated the effect of rabies virus infection on interleukin-1 alpha (IL-1 alpha) production and its receptors in mouse brain. Study of virus dissemination in the central nervous system (CNS) showed a massive infection of main brain structures from day 4 post infection (p.i.) up to the agony stage on day 6 p.i. At the same time, IL-1 alpha concentrations increased in cortical and hippocampal homogenates, whereas no change was detected in serum. In non-infected mice, IL-1 alpha binding sites were observed in the dentate gyrus, the cortex, the choroid plexus, the meninges and the anterior pituitary. During rabies virus infection, a striking decrease in IL-1 alpha binding sites was observed on day 4 p.i. with a complete disappearance on day 6 p.i., except in the pituitary gland where they remained at control level. In conclusion, concomitantly with the early rabid pathological signs, brain IL-1 alpha production and IL-1 alpha binding sites are specifically and significantly altered by brain viral proliferation. These results indicate that IL-1 alpha could be involved in the brain response to viral infection as a mediator and could participate in the genesis of the rabies pathogeny.


Subject(s)
Brain/metabolism , Interleukin-1/biosynthesis , Rabies/metabolism , Receptors, Interleukin-1/metabolism , Animals , Brain/pathology , Brain/virology , Cerebral Cortex/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Inbred BALB C , Rabies virus/physiology , Virus Replication
11.
J Cell Biol ; 128(5): 905-12, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7876313

ABSTRACT

Synapsin I is a synaptic vesicle-associated protein which inhibits neurotransmitter release, an effect which is abolished upon its phosphorylation by Ca2+/calmodulin-dependent protein kinase II (CaM kinase II). Based on indirect evidence, it was suggested that this effect on neurotransmitter release may be achieved by the reversible anchoring of synaptic vesicles to the actin cytoskeleton of the nerve terminal. Using video-enhanced microscopy, we have now obtained experimental evidence in support of this model: the presence of dephosphorylated synapsin I is necessary for synaptic vesicles to bind actin; synapsin I is able to promote actin polymerization and bundling of actin filaments in the presence of synaptic vesicles; the ability to cross-link synaptic vesicles and actin is specific for synapsin I and is not shared by other basic proteins; the cross-linking between synaptic vesicles and actin is specific for the membrane of synaptic vesicles and does not reflect either a non-specific binding of membranes to the highly surface active synapsin I molecule or trapping of vesicles within the thick bundles of actin filaments; the formation of the ternary complex is virtually abolished when synapsin I is phosphorylated by CaM kinase II. The data indicate that synapsin I markedly affects synaptic vesicle traffic and cytoskeleton assembly in the nerve terminal and provide a molecular basis for the ability of synapsin I to regulate the availability of synaptic vesicles for exocytosis and thereby the efficiency of neurotransmitter release.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Synapsins/metabolism , Synaptic Vesicles/metabolism , Actins/ultrastructure , Animals , Cattle , Cytoskeleton/ultrastructure , Energy Transfer , Microscopy, Fluorescence , Microscopy, Video , Phosphorylation , Prosencephalon/metabolism , Prosencephalon/ultrastructure , Protein Binding , Rabbits , Rats , Synaptic Vesicles/ultrastructure
12.
FEBS Lett ; 329(3): 301-5, 1993 Aug 30.
Article in English | MEDLINE | ID: mdl-8365471

ABSTRACT

Synapsin I is a nerve terminal phosphoprotein which interacts with synaptic vesicles and actin in a phosphorylation-dependent manner. By using fluorescence resonance energy transfer between purified components labeled with fluorescent probes, we now show that the binding of synapsin I to actin is a rapid phenomenon. Binding of synapsin I to actin can also be demonstrated when synaptic vesicles are present in the medium and appears to be modulated by ionic strength and synapsin I phosphorylation.


Subject(s)
Actins/metabolism , Synapsins/metabolism , Animals , Binding Sites , Cattle , Spectrometry, Fluorescence
13.
Eur J Pharmacol ; 245(2): 129-38, 1993 Apr 15.
Article in English | MEDLINE | ID: mdl-8491253

ABSTRACT

Rabies virus infection in man induces a series of clinical symptoms, some suggesting involvement of the central serotonergic system. The results of the present study show that, 5 days after rabies virus infection in rat, the total reversible high-affinity binding of [3H]5-HT in the hippocampus is not affected, suggesting that 5-HT1A binding is not altered. 5-HT1B sites identified by [125I]cyanopindolol binding are not affected in the cortex 3 and 5 days after the infection. Accordingly, the cellular inhibitory effect of trifluoromethylphenylpiperazine (TFMPP) on the [3H]acetylcholine-evoked release, presumably related to 5-HT1B receptor activity, is not modified 3 days after infection. In contrast, [3H]5-HT binding determined in the presence of drugs masking 5-HT1A, 5-HT1B and 5-HT1C receptors, is markedly (50%) reduced 3 days after the viral infection. These results suggest that 5-HT1D-like receptor subtypes may be affected specifically and at an early stage after rabies viral infection.


Subject(s)
Brain Chemistry/physiology , Rabies/metabolism , Receptors, Serotonin/metabolism , Acetylcholine/metabolism , Animals , Blotting, Northern , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Fluorescent Antibody Technique , Hippocampus/drug effects , Hippocampus/metabolism , In Vitro Techniques , Iodocyanopindolol , Male , Nucleic Acid Hybridization , Pindolol/analogs & derivatives , Pindolol/metabolism , Piperazines/metabolism , Potassium/pharmacology , Rats , Rats, Wistar , Synapses/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
14.
Mol Cell Probes ; 7(1): 1-5, 1993 Feb.
Article in English | MEDLINE | ID: mdl-7681151

ABSTRACT

The kinetics of viral RNA synthesis in different parts of the rat brain, infected with fixed or street rabies virus strains, is correlated with their anatomical neuronal connections with the masseter muscles, using hybridization with rabies cDNA probes. Viral RNA synthesis is first detected in the brain-stem and in the pons where the direct anatomical projection of the masseter muscle nervous arborization into the sensory and motor nuclei is located, through the trigeminus nerve. Rabies RNA detection is delayed in the other regions of the rat brain depending on the time course of virus transport from the trigeminal nuclei through multiple nervous connections.


Subject(s)
Axonal Transport , Brain/microbiology , DNA Probes , DNA/genetics , Masseter Muscle/microbiology , RNA, Messenger/biosynthesis , RNA, Viral/biosynthesis , Rabies virus/isolation & purification , Trigeminal Nerve/microbiology , Afferent Pathways/microbiology , Animals , Biomarkers , Masseter Muscle/innervation , Nucleic Acid Hybridization , Organ Specificity , RNA, Messenger/analysis , RNA, Viral/analysis , Rabies virus/genetics , Rats , Rats, Wistar
15.
J Physiol Paris ; 87(2): 117-22, 1993.
Article in English | MEDLINE | ID: mdl-8305899

ABSTRACT

Synapsin I is a neuron-specific phosphoprotein which binds to small synaptic vesicles and actin in a phosphorylation-dependent fashion. We have analyzed the ability of synapsin I to interact with actin monomers and filaments using purified proteins derivatized with fluorescent probes. Synapsin I accelerates the initial rate of actin polymerization and increases the final steady-state levels of polymerized actin. The fraction of total actin polymerized by synapsin I strongly depends on the synapsin I-actin ratio. We have visualized the actin-bundling activity of synapsin I using a non-perturbing method, video-enhanced microscopy of fluoresceinated synapsin I and actin filaments. Our findings suggest that synapsin I exerts a control on the physical characteristics of the cytoskeletal network of the nerve terminal and are consistent with the proposed role of synapsin I in mediating the interaction of synaptic vesicles with actin.


Subject(s)
Actins/physiology , Synapsins/physiology , Animals , Cattle , Fluorescein , Fluoresceins , Fluorometry , Microscopy/methods , Polymers/metabolism , Television
16.
J Gen Virol ; 72 ( Pt 5): 1191-4, 1991 May.
Article in English | MEDLINE | ID: mdl-2033395

ABSTRACT

Cultured human sensory neurons are directly susceptible to CVS rabies virus infection and produce virus yields of 10(5) p.f.u./ml; infection can persist for more than 20 days without any sign of c.p.e. The use of a compartmentalized two-chamber culture system, with access to either the cell soma or neuritic extensions, permitted the study of viral retrograde transport, which occurs at between 50 and 100 mm/day. Neurons of human origin were more susceptible to virus infection than rat neurons and the axonal transport of rabies virus was more efficient. Electron microscopy allowed virus transport and infection of human dorsal root ganglia neurons to be observed.


Subject(s)
Ganglia, Spinal/microbiology , Neurons, Afferent/microbiology , Rabies virus/isolation & purification , Axons/microbiology , Axons/ultrastructure , Biological Transport , Cells, Cultured , Fluorescent Antibody Technique , Ganglia, Spinal/ultrastructure , Humans , Kinetics , Microscopy, Electron , Neurons, Afferent/ultrastructure , Rabies virus/metabolism , Rabies virus/physiology , Virus Replication
17.
Antimicrob Agents Chemother ; 35(3): 572-4, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1674849

ABSTRACT

A noncompetitive N-methyl-D-aspartate (NMDA) antagonist, MK-801 (0.5 to 2.0 mM), inhibits rabies virus infection in rat primary cortical neurons, whereas the competitive NMDA antagonist AP5 has no effect. The results suggest that MK-801-mediated inhibition of rabies virus replication, although selective, is not operating through the high-affinity binding site mechanism.


Subject(s)
Dizocilpine Maleate/pharmacology , N-Methylaspartate/antagonists & inhibitors , Neurons/drug effects , Rabies virus/drug effects , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Cells, Cultured , Rabies/drug therapy , Rats , Virus Replication/drug effects
18.
Mol Cell Probes ; 4(3): 189-91, 1990 Jun.
Article in English | MEDLINE | ID: mdl-1696357

ABSTRACT

In an attempt to improve the sensitivity of the rabies genome hybridization test, PCR amplification was used following reverse transcription of rabies RNA extracted from infected brain. Presence of amplified DNA is demonstrated with either cDNA synthesized from the antigenomic primer or from antimessenger primer.


Subject(s)
Gene Amplification , Polymerase Chain Reaction , RNA, Viral/isolation & purification , Rabies virus/isolation & purification , Rabies/diagnosis , Animals , Base Sequence , Brain/microbiology , DNA/genetics , DNA, Viral/analysis , Mice , Molecular Sequence Data , RNA/genetics , RNA/isolation & purification , RNA, Messenger/genetics , RNA, Viral/genetics , Rabies/microbiology , Rabies virus/genetics
19.
J Virol Methods ; 28(1): 79-83, 1990 Apr.
Article in English | MEDLINE | ID: mdl-1693370

ABSTRACT

Rabies virus is a neurotropic agent which spreads in the CNS via axonal transport. Previous studies had shown that this axonal transport through the brain could be inhibited by stereotaxic administration of colchicine; however, this inhibition was reversible. We describe here a method to enhance the duration of this colchicine-mediated inhibition by delivering the drug continuously in the rat brain with osmotic pumps.


Subject(s)
Brain/microbiology , Colchicine/administration & dosage , Rabies virus/drug effects , Rabies/drug therapy , Animals , Axonal Transport/drug effects , Colchicine/therapeutic use , Infusion Pumps, Implantable , Male , Osmotic Pressure , Rabies/microbiology , Rats , Rats, Inbred Strains
20.
J Neuropathol Exp Neurol ; 48(6): 620-30, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2477505

ABSTRACT

The effect of colchicine, an inhibitor of axonal transport, on the spread of rabies virus in the central nervous system was investigated using Wistar rats. Colchicine was inoculated into the striatum at various times before and after inoculation of rabies virus into the same site. Rats were killed at various times after viral inoculation and the spread of rabies virus was monitored by rabies immunofluorescence of selected areas of brain. The most effective inhibitory effect was obtained by colchicine treatment applied two days before virus inoculation. Under these conditions, no fluorescent foci could be detected until day 3 post-infection whereas control rats exhibited infected cells as soon as two days post-infection. This inhibitory effect is reversible and the general consequence seems to be a delay in the rate of viral spread. However, five days after the virus challenge, some major brain areas were still partially preserved from infection (striatum, frontal cortex, pyriform cortex). Ten days after colchicine treatment, the microtubules have recovered their capacity to transport the virus. At the onset of paralysis, the general pattern of infection in brain sections from colchicine-treated rats was not significantly different from that of control rats. This inhibitory effect on the transport of rabies virus can be prolonged by administration of additional colchicine.


Subject(s)
Axonal Transport/drug effects , Brain/microbiology , Colchicine/pharmacology , Rabies virus/physiology , Rabies/prevention & control , Animals , Brain/drug effects , Colchicine/administration & dosage , Colchicine/therapeutic use , Male , Microscopy, Fluorescence , Peripheral Nerves/microbiology , Rabies virus/isolation & purification , Rats , Rats, Inbred Strains , Stereotaxic Techniques
SELECTION OF CITATIONS
SEARCH DETAIL