Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 18(1): 300, 2021 Dec 23.
Article in English | MEDLINE | ID: mdl-34949194

ABSTRACT

BACKGROUND: Atrial natriuretic peptide (ANP) secreted from atrial myocytes is shown to possess anti-inflammatory, anti-oxidant and immunomodulatory effects. The aim of this study is to assess the effect of ANP on bacterial lipopolysaccharide (LPS)-induced endotoxemia-derived neuroinflammation and cognitive impairment. METHODS: LPS (5 mg/kg) was given intraperitoneally to mice. Recombinant human ANP (rhANP) (1.0 mg/kg) was injected intravenously 24 h before and/or 10 min after LPS injection. Subdiaphragmatic vagotomy (SDV) was performed 14 days before LPS injection or 28 days before fecal microbiota transplantation (FMT). ANA-12 (0.5 mg/kg) was administrated intraperitoneally 30 min prior to rhANP treatment. RESULTS: LPS (5.0 mg/kg) induced remarkable splenomegaly and an increase in the plasma cytokines at 24 h after LPS injection. There were positive correlations between spleen weight and plasma cytokines levels. LPS also led to increased protein levels of ionized calcium-binding adaptor molecule (iba)-1, cytokines and inducible nitric oxide synthase (iNOS) in the hippocampus. LPS impaired the natural and learned behavior, as demonstrated by an increase in the latency to eat the food in the buried food test and a decrease in the number of entries and duration in the novel arm in the Y maze test. Combined prophylactic and therapeutic treatment with rhANP reversed LPS-induced splenomegaly, hippocampal and peripheral inflammation as well as cognitive impairment. However, rhANP could not further enhance the protective effects of SDV on hippocampal and peripheral inflammation. We further found that PGF mice transplanted with fecal bacteria from rhANP-treated endotoxemia mice alleviated the decreased protein levels of hippocampal polyclonal phosphorylated tyrosine kinase receptor B (p-TrkB), brain-derived neurotrophic factor (BDNF) and cognitive impairment, which was abolished by SDV. Moreover, TrkB/BDNF signaling inhibitor ANA-12 abolished the improving effects of rhANP on LPS-induced cognitive impairment. CONCLUSIONS: Our results suggest that rhANP could mitigate LPS-induced hippocampal inflammation and cognitive dysfunction through subdiaphragmatic vagus nerve-mediated gut microbiota-brain axis.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Brain-Gut Axis/drug effects , Cognitive Dysfunction/chemically induced , Endotoxins/antagonists & inhibitors , Gastrointestinal Microbiome/drug effects , Vagus Nerve/microbiology , Animals , Cognitive Dysfunction/psychology , Endotoxins/toxicity , Feces/microbiology , Inflammation Mediators , Injections, Intraperitoneal , Lipopolysaccharides/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/microbiology , Recombinant Proteins , Vagotomy
2.
J Neuroinflammation ; 17(1): 241, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32799901

ABSTRACT

BACKGROUND: The brain-gut-microbiota axis plays a role in the pathogenesis of stress-related disorders such as depression. In this study, we examined the effects of fecal microbiota transplantation (FMT) in mice with antibiotic-treated microbiota depletion. METHODS: The fecal microbiota was obtained from mice subjected to chronic social defeat stress (CSDS) and control (no CSDS) mice. FMT from these two groups was performed to antibiotic-treated mice. 16S rRNA analysis was performed to examine the composition of gut microbiota. Furthermore, the effects of subdiaphragmatic vagotomy in depression-like phenotypes after ingestion of microbes were examined. RESULTS: The ingestion of fecal microbiota from CSDS-susceptible mice resulted in an anhedonia-like phenotype, higher plasma levels of interleukin-6 (IL-6), and decreased expression of synaptic proteins in the prefrontal cortex (PFC) in antibiotic-treated mice but not in water-treated mice. 16S rRNA analysis suggested that two microbes (Lactobacillus intestinalis and Lactobacillus reuteri) may be responsible for the anhedonia-like phenotype in antibiotic-treated mice after FMT. Ingestion of these two microbes for 14 days led to depression- and anhedonia-like phenotypes, higher plasma IL-6 levels, and decreased expression of synaptic proteins in the PFC of antibiotic-treated mice. Interestingly, subdiaphragmatic vagotomy significantly blocked the development of behavioral abnormalities, elevation of plasma IL-6 levels, and downregulation of synaptic proteins in the PFC after ingestion of these two microbes. CONCLUSIONS: These findings suggest that microbiota depletion using an antibiotic cocktail is essential for the development of FMT-induced behavioral changes and that the vagus nerve plays a key role in behavioral abnormalities in antibiotic-treated mice after the ingestion of L. intestinalis and L. reuteri. Therefore, it is likely that the brain-gut-microbiota axis participates in the pathogenesis of depression via the vagus nerve.


Subject(s)
Anhedonia/drug effects , Anti-Bacterial Agents/pharmacology , Depression/microbiology , Lactobacillus , Limosilactobacillus reuteri , Vagus Nerve/microbiology , Animals , Depression/blood , Gastrointestinal Microbiome , Interleukin-6/blood , Mice , Motor Activity/drug effects , Stress, Psychological/blood , Stress, Psychological/microbiology
3.
Neuropharmacology ; 170: 108067, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32224131

ABSTRACT

There is accumulating evidence that certain gut microbes modulate brain chemistry and have antidepressant-like behavioural effects. However, it is unclear which brain regions respond to bacteria-derived signals or how signals are transmitted to distinct regions. We investigated the role of the vagus in mediating neuronal activation following oral treatment with Lactobacillus rhamnosus (JB-1). Male Balb/c mice were orally administered a single dose of saline or a live or heat-killed preparation of a physiologically active bacterial strain, Lactobacillus rhamnosus (JB-1). 165 min later, c-Fos immunoreactivity in the brain was mapped, and mesenteric vagal afferent fibre firing was recorded. Mice also underwent sub-diaphragmatic vagotomy to investigate whether severing the vagus prevented JB-1-induced c-Fos expression. Finally, we examined the ΔFosB response following acute versus chronic bacterial treatment. While a single exposure to live and heat-killed bacteria altered vagal activity, only live treatment induced rapid neural activation in widespread but distinct brain regions, as assessed by c-Fos expression. Sub-diaphragmatic vagotomy abolished c-Fos immunoreactivity in most, but not all, previously responsive regions. Chronic, but not acute treatment induced a distinct pattern of ΔFosB expression, including in previously unresponsive brain regions. These data identify that specific brain regions respond rapidly to gut microbes via vagal-dependent and independent pathways, and demonstrate that acute versus long-term exposure is associated with differential responses in distinct brain regions.


Subject(s)
Brain/metabolism , Brain/microbiology , Lacticaseibacillus rhamnosus/metabolism , Neurons/metabolism , Neurons/microbiology , Vagus Nerve/metabolism , Vagus Nerve/microbiology , Administration, Oral , Animals , Male , Mice , Mice, Inbred BALB C , Vagotomy/trends , Vagus Nerve/surgery
4.
Neurosci Lett ; 662: 98-104, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-28993208

ABSTRACT

OBJECTIVE: The pathogenesis of sepsis associated encephalopathy (SAE) remains poorly understood. Vagus nerve plays an important role in gut-microbiota-brain axis. This study aimed to investigate whether vague nerve is a key mediator of the impact of intestinal microbiota on SAE. METHODS: Male rats were randomly divided into four groups (n=20): SHAM (SH) group, lipopolysaccharide (LPS) group, fecal microbiota transplantation (FMT) +LPS group, and vagotomy (VGX)+LPS+FMT group. The left cervical vagotomy was performed 30min before LPS administration in LPS+FMT+VGX group. LPS+ FMT and LPS+FMT+VGX groups received nasogastric infusion of feces from healthy donor three times a day. Fecal samples were collected every two days to monitor changes in microbiota composition by 16S rDNA analysis. Brain function was evaluated by behavioral tests and EEG. The levels of tumor necrosis factor alpha (TNF-α), interleukin (IL)-1ß, IL-6, IL-10 in brain cortex were detected by ELISA. The expression of Iba-1 in brain cortex was assessed by immunohistochemistry and Western blot analysis. RESULTS: Significant modification of microbiota composition, characterized by a profound increase of commensals in the Firmicutes phylum and depletion of opportunistic organisms in the Proteobacteria phylum, was observed in FMT groups compared to LPS group. Furthermore, we identified a reconstituted bacterial community enriched in Firmicutes and depleted of Proteobacteria. In both FMT groups the diversity of the fecal microbiota and the microbiota composition were similar to SH group. LPS mice treated with FMT demonstrated a better spatial memory and less EEG abnormalities, significantly attenuated levels of IL-1ß, IL-6, TNF-α, and decreased number of Iba-1 positive microglia in the cortex, but these beneficial effects of FMT were reversed by VGX. CONCLUSIONS: FMT can change intestinal microbiota in sepsis patients, and vagus nerve is a key mediator between intestinal microbiota and SAE. These findings suggest that FMT and vagus nerve are potential therapy targets for treating SAE.


Subject(s)
Gastrointestinal Microbiome/physiology , Sepsis-Associated Encephalopathy/microbiology , Sepsis-Associated Encephalopathy/physiopathology , Vagus Nerve/physiopathology , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cytokines/metabolism , Feces/microbiology , Hippocampus/metabolism , Lipopolysaccharides/pharmacology , Male , Memory , Microglia/metabolism , Rats, Sprague-Dawley , Sepsis-Associated Encephalopathy/psychology , Spatial Learning , Vagus Nerve/microbiology
5.
J Neuroimmunol ; 257(1-2): 36-45, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23481698

ABSTRACT

Vagal activation can reduce inflammation and disease activity in various animal models of intestinal inflammation via the cholinergic anti-inflammatory pathway. In the current model of this pathway, activation of descending vagal efferents is dependent on a signal initiated by stimulation of vagal afferents. However, little is known about how vagal afferents are activated, especially in the context of subclinical or clinical pathogenic bacterial infection. To address this question, we first determined if selective lesions of capsaicin-sensitive vagal afferents altered c-Fos expression in the nucleus of the solitary tract (nTS) after mice were inoculated with either Campylobacter jejuni or Salmonella typhimurium. Our results demonstrate that the activation of nTS neurons by intraluminal pathogenic bacteria is dependent on intact, capsaicin sensitive vagal afferents. We next determined if inflammatory mediators could cause the observed increase in c-Fos expression in the nTS by a direct action on vagal afferents. This was tested by the use of single-cell calcium measurements in cultured vagal afferent neurons. We found that tumor necrosis factor alpha (TNFα) and lipopolysaccharide (LPS) directly activate cultured vagal afferent neurons and that almost all TNFα and LPS responsive neurons were sensitive to capsaicin. We conclude that activation of the afferent arm of the parasympathetic neuroimmune reflex by pathogenic bacteria in the gut is dependent on capsaicin sensitive vagal afferent neurons and that the release of inflammatory mediators into intestinal tissue can be directly sensed by these neurons.


Subject(s)
Capsaicin/pharmacology , Gastrointestinal Tract/microbiology , Neurons, Afferent/microbiology , Vagus Nerve/microbiology , Animals , Campylobacter Infections/microbiology , Campylobacter Infections/pathology , Colony Count, Microbial , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/pathology , Humans , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , Neurons, Afferent/drug effects , Neurons, Afferent/pathology , Rats , Rats, Sprague-Dawley , Salmonella Infections/microbiology , Salmonella Infections/pathology , Vagus Nerve/drug effects , Vagus Nerve/pathology
6.
Surg Neurol ; 66(1): 94-9; discussion 99, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16793457

ABSTRACT

BACKGROUND: CVJ tuberculosis is a described entity requiring challenging ways of management. Severe neck pain, causing restricted neck movements and torticollis, is a characteristic presentation of neurologically asymptomatic suboccipital Pott's disease. CASE DESCRIPTION: Two patients with unusual CVJ tuberculosis form the basis for the present communication. The first patient presented with tubercular otitis media, causing progressive erosion of the petrous part of temporal bone, and destruction of the occipital condyle, along with the lateral mass of atlas, leading to CVJ instability. This is a first report of such a presentation, according to our knowledge. Detailed bony architectural destruction demonstrable on CT scan has been described. The second patient, with CVJ tuberculosis, presented with skull base syndrome and with multiple cranial nerve palsies. Both patients were managed without surgical intervention and showed clinical and radiological recovery. CONCLUSION: In such patients with unusual clinical presentations, histopathologic examination is necessary to arrive at a correct diagnosis. The management of patients with tubercular involvement of CVJ remains controversial. In the present communication, both the patients were managed successfully with full dose of antitubercular drugs and immobilization.


Subject(s)
Axis, Cervical Vertebra/pathology , Cervical Atlas/pathology , Occipital Bone/pathology , Otitis Media/complications , Skull Base/pathology , Tuberculosis, Spinal/diagnosis , Adult , Axis, Cervical Vertebra/diagnostic imaging , Axis, Cervical Vertebra/microbiology , Cervical Atlas/diagnostic imaging , Cervical Atlas/microbiology , Cranial Nerve Diseases/diagnosis , Cranial Nerve Diseases/microbiology , Cranial Nerve Diseases/physiopathology , Disease Progression , Ear, Middle/diagnostic imaging , Ear, Middle/microbiology , Ear, Middle/pathology , Early Diagnosis , Female , Humans , Hypoglossal Nerve/microbiology , Hypoglossal Nerve/pathology , Hypoglossal Nerve/physiopathology , India , Male , Middle Aged , Neck Pain/diagnosis , Neck Pain/microbiology , Neck Pain/physiopathology , Occipital Bone/diagnostic imaging , Occipital Bone/microbiology , Otitis Media/diagnosis , Otitis Media/microbiology , Skull Base/diagnostic imaging , Skull Base/microbiology , Temporal Bone/diagnostic imaging , Temporal Bone/microbiology , Temporal Bone/pathology , Tomography, X-Ray Computed , Tuberculosis, Spinal/physiopathology , Tuberculosis, Spinal/therapy , Vagus Nerve/microbiology , Vagus Nerve/pathology , Vagus Nerve/physiopathology
8.
Science ; 263(5144): 232-4, 1994 Jan 14.
Article in English | MEDLINE | ID: mdl-8284675

ABSTRACT

The vagus nerve exerts a profound influence on the heart, regulating the heart rate and rhythm. An extensive vagal innervation of the cardiac ventricles and the central origin and extent of this innervation was demonstrated by transynaptic transport of pseudorabies virus with a virulent and two attenuated pseudorabies viral strains. The neurons that innervate the ventricles are numerous, and their distribution within the nucleus ambiguus and dorsal motor nucleus of the vagus is similar to that of neurons innervating other cardiac targets, such as the sino-atrial node. These data provide a neuroanatomical correlate to the physiological influence of the vagus nerve on ventricular function.


Subject(s)
Heart Ventricles/innervation , Herpesvirus 1, Suid/physiology , Medulla Oblongata/anatomy & histology , Vagus Nerve/anatomy & histology , Animals , Brain Mapping , Heart Ventricles/microbiology , Herpesvirus 1, Suid/pathogenicity , Interneurons/cytology , Medulla Oblongata/microbiology , Motor Neurons/cytology , Neural Pathways , Rats , Rats, Wistar , Vagus Nerve/microbiology , Virulence
9.
J Neurosci ; 13(2): 685-702, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8381171

ABSTRACT

We examined the responses of astrocytes, ramified microglia, and brain macrophages to CNS neuronal infection with virulent or attenuated strains of a swine alpha herpesvirus (pseudorabies virus, PRV). After PRV inoculation of the rat stomach or pancreas, the temporal course of viral replication and induced pathology of infected neurons were assessed in the dorsal motor nucleus of the vagus (DMV) and amygdala using an antiserum generated against PRV. Specific monoclonal antibodies against glial fibrillary acidic protein (GFAP), OX42, and ED1 and morphological criteria were used to classify non-neuronal cells. Both PRV strains infected DMV and motor neurons and then passed transneuronally to infect brainstem neurons that innervate the DMV. However, the onset of neuronal infection produced by the attenuated strain occurred approximately 20 hr later than infection with the virulent strain. Animals infected with the attenuated strain also survived longer, permitting transneuronal passage of virus into forebrain areas of the visceral neuraxis. Neuronal infection with both PRV strains produced consistent alterations in astrocytes, ramified microglia, and brain macrophages that correlated spatially and temporally with progressive stages of viral replication and neuronal pathology. Early stages of infection were characterized by increases in immunoreactivity for astrocytic GFAP and microglial OX42 that preceded overt signs of neuronal pathology. At later stages, GFAP immunoreactivity decreased dramatically in focal areas of neuronal infection while OX42 immunoreactivity continued to increase. Subsequently, ED1-immunoreactive brain macrophages infiltrated these infected areas. Double immunocytochemical labeling demonstrated that some astrocytes and brain macrophages were immunopositive for viral antigens but ramified microglia were not. The responses of glia and brain macrophages are consistent with a proposed role in restricting extracellular spread of virus by isolating or phagocytosing infected cells. These phenomena may contribute to the specific transneuronal transport exhibited by PRV.


Subject(s)
Astrocytes/pathology , Brain/pathology , Macrophages/pathology , Motor Neurons/microbiology , Neuroglia/pathology , Pseudorabies/pathology , Animals , Antibodies, Monoclonal , Antigens, Surface/analysis , Astrocytes/chemistry , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/analysis , Herpesvirus 1, Suid/isolation & purification , Herpesvirus 1, Suid/physiology , Male , Rats , Rats, Sprague-Dawley , Receptors, Complement 3b/analysis , Receptors, Complement 3b/immunology , Vagus Nerve/cytology , Vagus Nerve/microbiology , Virus Replication
10.
Proc Natl Acad Sci U S A ; 88(9): 3852-6, 1991 May 01.
Article in English | MEDLINE | ID: mdl-1850838

ABSTRACT

A crucial event in the pathogenesis of systemic enteric virus infections is entry of virus into the nervous system. Whether enteric virus spreads from the intestinal tract to the central nervous system through nerves or through the bloodstream was examined using a serotype 3 reovirus strain. After peroral inoculation of newborn mice with reovirus, serial histologic sections of small intestine, brain and spinal cord were prepared and stained by immunoperoxidase to detect viral antigen. Three days after inoculation, viral antigen was observed in mononuclear cells of ileal Peyer's patches and in neurons of the adjacent myenteric plexus. Infection first appeared in the central nervous system 1-2 days later in neurons of the dorsal motor nucleus of the vagus nerve. Endothelial cells, meninges, choroid plexus, hypothalamus, and area postrema were not infected, indicating neural rather than bloodborne spread from the intestine. Staining of neurons in the dorsal motor nucleus of the vagus nerve depended on the route of virus inoculation and was independent of the amount of virus in the bloodstream. These results demonstrate that an enteric virus entering a host from the intestinal lumen can spread to the central nervous system through nerve fiber innervating the intestine.


Subject(s)
Intestines/microbiology , Reoviridae Infections/microbiology , Reoviridae/growth & development , Animals , Antigens, Viral/analysis , Brain/microbiology , Brain Stem/microbiology , Mice , Myenteric Plexus/microbiology , Peyer's Patches/microbiology , Reoviridae/immunology , Vagus Nerve/microbiology
11.
Neuroscience ; 42(1): 261-74, 1991.
Article in English | MEDLINE | ID: mdl-1650433

ABSTRACT

The recent introduction of live viruses as intra-axonal tracing agents has raised questions concerning which central neurons are transneuronally labelled after application of the virus to peripheral organs or peripheral nerves. Since the central connections of the vagus nerve have been well described using conventional neuronal tracing agents, we chose to inject Herpes Simplex Virus Type 1 into the cervical vagus of the rat. After survival times of up to 3 days the rat brains were processed immunohistochemically using a polyclonal antiserum against herpes simplex virus. Two days after injection of the virus we observed viral antigen in the area postrema and in the nucleus tractus solitarius and the dorsal motor nucleus of the vagus (dorsal vagal complex), principally ipsilaterally. At this survival time the viral antigen in the dorsal vagal complex was largely confined to glial cells. After 3 days the viral antigen was localized both in glia and in nerve cells within the dorsal vagal complex and in brain regions previously demonstrated, using conventional tracing procedures, to contain neurons with axonal projections to the dorsal vagal complex. This was true for medullary, pontine, midbrain and hypothalamic regions and for telencephalic regions including the amygdala, the bed nucleus of the stria terminalis, and the insular and medial frontal cortices. Many of the nerve cells containing viral antigen were displayed in a Golgi-like manner, with excellent visualization of the dendritic tree. Axonal processes, in contrast, were not visualized. We used co-localization studies to confirm previous findings concerning monoamine neurotransmitter-related antigens present in medullary and pontine neurons projecting to the dorsal vagal complex. After 3 days there were many Herpes Simplex Virus Type 1-containing glial cells along the intra-medullary course of the vagal rootlets. However, no viral antigen was found in brain regions containing neurons whose axons pass through the region of glial cell-labelled rootlets. Glial cells containing viral antigen were particularly numerous in brain regions known to receive an input from neurons in the area postrema and the dorsal vagal complex. Taken together with our observation concerning the early appearance of viral antigen within glial cells in the dorsal vagal complex, this suggests that when the virus reaches the axon terminal portion it is transferred to nearby glial cells and possibly enters central neurons by way of these structures.


Subject(s)
Axons/microbiology , Neurons, Afferent/microbiology , Neurons/microbiology , Simplexvirus/metabolism , Stilbamidines , Vagus Nerve/microbiology , Amidines , Animals , Antibodies, Viral/analysis , Fluorescent Dyes , Immunohistochemistry , Medulla Oblongata/anatomy & histology , Medulla Oblongata/microbiology , Neuroglia/immunology , Raphe Nuclei/anatomy & histology , Raphe Nuclei/microbiology , Rats
12.
J Comp Pathol ; 103(3): 301-6, 1990 Oct.
Article in English | MEDLINE | ID: mdl-2175319

ABSTRACT

The MacIntyre strain of Herpes simplex virus type 1 was inoculated intraperitoneally into young male mice. Immunohistochemical detection of the virus antigen demonstrated the passage of virus from the gastro-intestinal myenteric plexus towards the brain and its distribution within the areas of brain related to the principal nuclei of the vagus nerve. Selective trapping of the virus in the autonomic nerves and subsequent localization of antigen behind the blood-brain barrier may prevent therapeutic effects of agents tested in this model.


Subject(s)
Abdomen/microbiology , Brain/microbiology , Simplexvirus/growth & development , Vagus Nerve/microbiology , Animals , Male , Mice , Mice, Inbred BALB C
14.
J Virol ; 52(1): 300-5, 1984 Oct.
Article in English | MEDLINE | ID: mdl-6090708

ABSTRACT

One hundred and fifteen isolates of herpes simplex virus were recovered from parallel explant cultures of trigeminal and vagus ganglia and trigeminal nerve roots derived from 20 unselected human cadavers. Restriction enzyme patterns of strains recovered from 18 of 20 individuals could be differentiated from individual to individual, although all isolates from a single host were identical. Isolates from two individuals differed among themselves in the number and location of certain restriction enzyme sites.


Subject(s)
Ganglia/microbiology , Simplexvirus/isolation & purification , Trigeminal Nerve/microbiology , Vagus Nerve/microbiology , Base Sequence , Cloning, Molecular , DNA Restriction Enzymes , DNA, Viral/isolation & purification , Genes, Viral , Humans , Simplexvirus/classification , Simplexvirus/genetics , Species Specificity
15.
J Gen Virol ; 43(1): 151-71, 1979 Apr.
Article in English | MEDLINE | ID: mdl-225415

ABSTRACT

Analysis of the infected cell polypeptides and the DNA restriction profiles of 31 HSV-1 isolates from the trigeminal, superior cervical and vagus ganglia from 17 individuals (12 U.S.A., 2 Japanese, 3 Norwegian) could be classified as 15 different virus strains. With the exception of the three Norwegian isolates which gave identical profiles, virus isolates from the ganglia of different individuals could all be distinguished from one another. In contrast virus isolates from the trigeminal, superior cervical and vagus ganglia of the same individual, or virus isolates from the left and right ganglia of the same individual or multiple isolates from different explants of a single ganglion were indistinguishable. In conclusion, a single virus strain infects each individual initially and virus descended from this event subsequently infects and becomes latent in different cells of the same ganglion as well as in different ganglia.


Subject(s)
DNA, Viral/analysis , Ganglia, Autonomic/microbiology , Nodose Ganglion/microbiology , Peptides/analysis , Simplexvirus/analysis , Trigeminal Ganglion/microbiology , Trigeminal Nerve/microbiology , Vagus Nerve/microbiology , DNA Restriction Enzymes/metabolism , Humans , Neck , Simplexvirus/growth & development , Viral Proteins/analysis , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...