Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Handb Exp Pharmacol ; 239: 269-287, 2017.
Article in English | MEDLINE | ID: mdl-28035535

ABSTRACT

A growing body of preclinical and clinical evidence supports a relationship between the complexity and diversity of the microorganisms that inhabit our gut (human gastrointestinal microbiota) and health status. Under normal homeostatic conditions this microbial population helps maintain intestinal peristalsis, mucosal integrity, pH balance, immune priming and protection against invading pathogens. Furthermore, these microbes can influence centrally regulated emotional behaviour through mechanisms including microbially derived bioactive molecules (amino acid metabolites, short-chain fatty acids, neuropeptides and neurotransmitters), mucosal immune and enteroendocrine cell activation, as well as vagal nerve stimulation.The microbiota-gut-brain axis comprises a dynamic matrix of tissues and organs including the brain, autonomic nervous system, glands, gut, immune cells and gastrointestinal microbiota that communicate in a complex multidirectional manner to maintain homeostasis and resist perturbation to the system. Changes to the microbial environment, as a consequence of illness, stress or injury, can lead to a broad spectrum of physiological and behavioural effects locally including a decrease in gut barrier integrity, altered gut motility, inflammatory mediator release as well as nociceptive and distension receptor sensitisation. Centrally mediated events including hypothalamic-pituitary-adrenal (HPA) axis, neuroinflammatory events and neurotransmitter systems are concomitantly altered. Thus, both central and peripheral pathways associated with pain manifestation and perception are altered as a consequence of the microbiota-gut-brain axis imbalance.In this chapter the involvement of the gastrointestinal microbiota in visceral pain is reviewed. We focus on the anatomical and physiological nodes whereby microbiota may be mediating pain response, and address the potential for manipulating gastrointestinal microbiota as a therapeutic target for visceral pain.


Subject(s)
Abdominal Pain/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/innervation , Gastrointestinal Tract/microbiology , Visceral Pain/microbiology , Visceral Pain/physiopathology , Abdominal Pain/physiopathology , Abdominal Pain/therapy , Animals , Brain/physiopathology , Host-Pathogen Interactions , Humans , Neural Pathways/microbiology , Neural Pathways/physiopathology , Pain Perception , Pain Threshold , Probiotics/therapeutic use , Visceral Pain/therapy
2.
J Neurosci ; 32(6): 1920-31, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22323705

ABSTRACT

Food intake in the nematode Caenorhabditis elegans requires two distinct feeding motions, pharyngeal pumping and isthmus peristalsis. Bacteria, the natural food of C. elegans, activate both feeding motions (Croll, 1978; Horvitz et al., 1982; Chiang et al., 2006). The mechanisms by which bacteria activate the feeding motions are largely unknown. To understand the process, we studied how serotonin, an endogenous pharyngeal pumping activator whose action is triggered by bacteria, activates feeding motions. Here, we show that serotonin, like bacteria, activates overall feeding by activating isthmus peristalsis as well as pharyngeal pumping. During active feeding, the frequencies and the timing of onset of the two motions were distinct, but each isthmus peristalsis was coupled to the preceding pump. We found that serotonin activates the two feeding motions mainly by activating two separate neural pathways in response to bacteria. For activating pumping, the SER-7 serotonin receptor in the MC motor neurons in the feeding organ activated cholinergic transmission from MC to the pharyngeal muscles by activating the Gsα signaling pathway. For activating isthmus peristalsis, SER-7 in the M4 (and possibly M2) motor neuron in the feeding organ activated the G(12)α signaling pathway in a cell-autonomous manner, which presumably activates neurotransmission from M4 to the pharyngeal muscles. Based on our results and previous calcium imaging of pharyngeal muscles (Shimozono et al., 2004), we propose a model that explains how the two feeding motions are separately regulated yet coupled. The feeding organ may have evolved this way to support efficient feeding.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Feeding Behavior/physiology , Motor Neurons/physiology , Receptors, Serotonin/physiology , Serotonin/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Eating/physiology , Gene Knockdown Techniques , Motor Neurons/microbiology , Mutation/physiology , Neural Pathways/microbiology , Neural Pathways/physiology , Peristalsis/physiology , Pharyngeal Muscles/microbiology , Pharyngeal Muscles/physiology
3.
Med Hypotheses ; 68(5): 1059-64, 2007.
Article in English | MEDLINE | ID: mdl-17113237

ABSTRACT

Brain structure in health is a dynamic energized equation incorporating chemistry, neuronal structure, and circuitry components. The chemistry "piece" is represented by multiple neurotransmitters such as Acetylcholine, Serotonin, and Dopamine. The neuronal structure "piece" incorporates synapses and their connections. And finally circuits of neurons establish "architectural blueprints" of anatomic wiring diagrams of the higher order of brain neuron organizations. In Alzheimer's disease, there are progressive losses in all of these components. Brain structure crumbles. The deterioration in Alzheimer's is ordered, reproducible, and stepwise. Drs. Braak and Braak have described stages in the Alzheimer disease continuum. "Progressions" through Braak Stages benchmark "Regressions" in Cognitive function. Under the microscope, the Stages of Braak commence in brain regions near to the hippocampus, and over time, like a tsunami wave of destruction, overturn healthy brain regions, with neurofibrillary tangle damaged neurons "marching" through the temporal lobe, neocortex and occipital cortex. In effect the destruction ascends from the limbic regions to progressively destroy the higher brain centers. Rabies infection also "begins low and finishes high" in its wave of destruction of brain tissue. Herpes Zoster infections offer the paradigm of clinical latency of infection inside of nerves before the "marching commences". Varicella Zoster virus enters neurons in the pediatric years. Dormant virus remains inside the neurons for 50-80 years, tissue damage late in life (shingles) demonstrates the "march of the infection" down neural pathways (dermatomes) as linear areas of painful blisters loaded with virus from a childhood infection. Amalgamation of Zoster with Rabies models produces a hybrid model to explain all of the Braak Stages of Alzheimer's disease under a new paradigm, namely "Alzheimer's neuroborreliosis" in which latent Borrelia infections ascend neural circuits through the hippocampus to the higher brain centers, creating a trail of neurofibrillary tangle injured neurons in neural circuits of cholinergic neurons by transsynaptic transmission of infection from nerve to nerve.


Subject(s)
Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Borrelia Infections/transmission , Borrelia/pathogenicity , Models, Neurological , Neural Pathways/microbiology , Disease Progression , Humans
4.
Neuroscience ; 57(4): 1007-25, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8309541

ABSTRACT

Several neurotropic viruses enter the brain after peripheral inoculation and spread transneuronally along pathways known to be connected to the initial site of entry. In this study, the pathways utilized by two such viruses, herpes simplex virus type 1 and mouse hepatitis virus strain JHM, were compared using in situ hybridization following inoculation into either the nasal cavity or the main olfactory bulb of the mouse. The results indicate that both viruses spread to infect a unique and only partially overlapping set of connections of the main olfactory bulb. Both quantitative and qualitative differences were observed in the patterns of infection of known primary and secondary main olfactory bulb connections. Using immunohistochemistry for tyrosine hydroxylase combined with in situ hybridization, it was shown that only herpes simplex virus infected noradrenergic neurons in the locus coeruleus. In contrast, both viruses infected dopaminergic neurons in the ventral tegmental area, although mouse hepatitis virus produced a more widespread infection in the A10 group, as well as infecting A8 and A9. The results suggest that differential virus uptake in specific neurotransmitter systems contributes to the pattern of viral spread, although other factors, such as differences in access to particular synapses on infected cells and differences in the distribution of the cellular receptor for the two viruses, are also likely to be important. The data show that neural tracing with different viruses may define unique neural pathways from a site of inoculation. The data also demonstrate that two viruses can enter the brain via the olfactory system and localize to different structures, suggesting that neurological diseases involving disparate regions of the brain could be caused by different viruses, even if entry occurred at a common site.


Subject(s)
Hepatitis Viruses/physiology , Olfactory Bulb/microbiology , Simplexvirus/physiology , Animals , Brain/microbiology , Dopamine/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Mice , Neural Pathways/microbiology , Neurons/microbiology , Norepinephrine/metabolism
5.
Adv Exp Med Biol ; 342: 333-8, 1993.
Article in English | MEDLINE | ID: mdl-8209750

ABSTRACT

Four-week-old Wistar rats were inoculated with HEV by different routes. Animals died of encephalitis after intraperitoneal (i.p.), subcutaneous (s.c.) and intravenous (i.v.) as well as intracerebral (i.c.) and intranasal (i.n.) inoculation. However when inoculated subcutaneously, rats died a few days earlier than those inoculated i.p. and i.v., suggesting that the virus might be transmitted to the central nervous system (CNS) by the neuronal route rather than by blood stream. Rats which were inoculated subcutaneously at the site of the neck (group A) began to die on day 4 p.i., a few days earlier than animals inoculated in the foot pad of the right leg (group B). On day 2 and 3 after inoculation, the virus titer in the brain was higher in group A, but group B animals showed higher virus titers in the lumber region of spinal cord than group A animals. In order to follow the virus spread from the peripheral nerve to the brain, the virus was inoculated into the sciatic nerve of rats. The inoculated rats developed clinical signs on day 4 and began to die on day 6. On day 2, virus was detected in the posterior half of the spinal cord and migrated toward the anterior half and in the brain where it was present on day 3. The highest virus titers in the brain were recorded on day 4 to 6, meanwhile the virus titers in the spinal cord tend to decrease. By immunohistochemical study, antigen positive neurons were found in the spinal cord and brain on day 4.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cerebral Cortex/microbiology , Coronavirus/physiology , Encephalomyelitis/microbiology , Neural Pathways/microbiology , Spinal Cord/microbiology , Administration, Intranasal , Animals , Antigens, Viral/analysis , Brain , Coronavirus/isolation & purification , Injections , Injections, Intraperitoneal , Injections, Intravenous , Injections, Subcutaneous , Neurons/microbiology , Rats , Rats, Wistar , Sciatic Nerve/microbiology , Time Factors
6.
J Infect Dis ; 166(4): 747-52, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1326581

ABSTRACT

A transgenic mouse model was used to address an unsolved question in the pathogenesis of poliomyelitis: how poliovirus invades the central nervous system (CNS). LD50 values for intramuscular and intracerebral inoculation of poliovirus in transgenic mice expressing poliovirus receptors (TgPVR mice) were similar. After intramuscular inoculation with poliovirus, paralysis was observed first in the inoculated limb. In contrast, localization of initial paralysis to the inoculated limb was not observed in normal mice inoculated intramuscularly with the mouse-adapted P2/Lansing poliovirus strain. After intramuscular inoculation, infectious poliovirus was first detected in the inferior segment of the spinal cord, then in the superior spinal cord and the brain. Sciatic nerve transection blocked poliovirus spread to the spinal cord after inoculation into the hindlimb footpad of TgPVR mice. These results demonstrate that in TgPVR mice, poliovirus spreads from muscle to the CNS through nerve pathways and that expression of the poliovirus receptor plays an important role in viral spread by this route.


Subject(s)
Central Nervous System/microbiology , Muscles/microbiology , Poliomyelitis/microbiology , Poliovirus/pathogenicity , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Neural Pathways/microbiology , Poliomyelitis/physiopathology , Sciatic Nerve/surgery
7.
Brain Pathol ; 1(1): 6-10, 1990 Sep.
Article in English | MEDLINE | ID: mdl-1669695

ABSTRACT

Cumulative evidence suggests that varicella-zoster virus (VZV) can infect walls of CNS arteries, causing stroke in man. We review observations relating infection with this neurotropic virus to the development of arteritis in the CNS and note evidence supporting the hypothesis that VZV spreads from ganglionic reactivation sites to the arterial wall by neural pathways. Problems relating to the pathogenesis of arteritis and experimental approaches to their solution are suggested.


Subject(s)
Arteritis/microbiology , Cerebral Arteries/microbiology , Cerebrovascular Disorders/microbiology , Herpes Zoster/complications , Adult , Aged , Aged, 80 and over , Animals , Animals, Domestic/microbiology , Arteritis/pathology , Cerebral Arteries/pathology , Cerebrovascular Disorders/pathology , Disease Models, Animal , Female , Giant Cell Arteritis/microbiology , Giant Cell Arteritis/pathology , Herpesviridae Infections/complications , Herpesviridae Infections/veterinary , Humans , Male , Middle Aged , Neural Pathways/microbiology
8.
J Neurosci ; 10(6): 1974-94, 1990 Jun.
Article in English | MEDLINE | ID: mdl-2162388

ABSTRACT

Uptake, replication, and transneuronal passage of a swine neurotropic herpesvirus (pseudorabies virus, PRV) was evaluated in the rat CNS. PRV was localized in neural circuits innervating the tongue, stomach, esophagus and eye with light microscopic immunohistochemistry. In each instance, the distribution of PRV-immunoreactive neurons was entirely consistent with that observed following injection of cholera toxin-horseradish peroxidase conjugate (CT-HRP). Injections of the tongue resulted in retrograde transport of PRV and CT-HRP to hypoglossal motor neurons, while preganglionic neurons in the dorsal motor vagal nucleus or somatic motor neurons in the nucleus ambiguus were labeled following injections of the stomach or esophagus, respectively. At longer times after infection, viral antigens were found in astrocytes adjacent to infected neurons and their efferent axons and second-order neuron labeling became apparent. The distribution of second-order neurons was also entirely dependent upon the site of PRV injection. Following tongue injection, second-order neurons were observed in the trigeminal complex, the brain-stem tegmentum and in monoaminergic cell groups. Injection of the stomach or esophagus led to second-order neuron labeling confined to distinct subdivisions of the neucleus of the solitary tract and monoaminergic cell groups. Comparative quantitative analysis of the number of PRV immunoreactive neurons present in the diencephalon and brain stem following injection of virus into both the eye and stomach musculature of the same animal demonstrated that retrograde transport of PRV from the viscera was more efficient and occurred at a much faster rate than anterograde transport of virus. These data demonstrate projection-specific transport of PRV in the nervous system and provide further insight into the means through which this neurotropic virus infects the nervous system.


Subject(s)
Central Nervous System/microbiology , Herpesvirus 1, Suid/physiology , Neurons/physiology , Animals , Brain Stem/microbiology , Esophagus/microbiology , Injections , Male , Neural Pathways/microbiology , Neurons/microbiology , Rats , Rats, Inbred Strains , Stomach/microbiology , Time Factors , Tongue/microbiology , Virus Replication
9.
J Microsc ; 141(Pt 1): 55-67, 1986 Jan.
Article in English | MEDLINE | ID: mdl-3007764

ABSTRACT

This investigation is concerned with the possibility of identifying viral DNA using the in situ DNA hybridization method in methylmethacrylate-embedded material. As an experimental model we chose viral labyrinthitis produced by intranasal infection of the mouse with pseudorabies virus. Fixation and embedding methods specially adapted to this procedure and bony histology preparation technique (specimens by grinding or micromilling) made it possible to identify viral DNA directly morphologically and virologically in the inner ear. Quantitative microphotometric analyses of trans-sagittal sections of the entire skull after in situ DNA hybridization are presented and discussed here as an explicit method of investigating the path of distribution of viral DNA in the brain and the inner ear.


Subject(s)
DNA, Viral/analysis , Ear, Inner/microbiology , Herpesvirus 1, Suid/genetics , Nucleic Acid Hybridization , Animals , Autoradiography , Brain/microbiology , Brain/pathology , Cochlea/microbiology , Cochlea/pathology , Histological Techniques , Labyrinthitis/microbiology , Labyrinthitis/pathology , Methylmethacrylates , Mice , Mice, Inbred BALB C , Neural Pathways/microbiology , Neural Pathways/pathology , Pseudorabies/microbiology
10.
J Neurol Sci ; 64(2): 213-7, 1984 May.
Article in English | MEDLINE | ID: mdl-6379105

ABSTRACT

An immunohistological study of viral antigen (VA) in the brain was carried out in mice which had been infected with herpes simplex type 1 virus (HSV) in the skin of the face. In 77% of the mice with VA in the brain the olfactory system as well as the trigeminal system/brainstem was affected. The remaining 23% had VA in the trigeminal system/brainstem only. Eye swab cultures yielded HSV from all mice with VA in the olfactory system. The ease of access of virus infecting the face to the olfactory system shown in this model may have implications for human infections.


Subject(s)
Central Nervous System/microbiology , Encephalitis/etiology , Herpes Simplex , Olfactory Pathways/microbiology , Skin Diseases, Infectious/etiology , Trigeminal Nerve/microbiology , Animals , Facial Dermatoses/transmission , Female , Herpes Simplex/transmission , Histocytochemistry , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Neural Pathways/microbiology
12.
Arkh Patol ; 40(3): 20-6, 1978.
Article in Russian | MEDLINE | ID: mdl-206238

ABSTRACT

Electron microscopic investigation of the brains and lumbar spinal cords of adult albino mice infected with Powassan virus was carried out. Virus particles were found within all parts of neurons (perikarya, dendrites, axon), as well as within synaptic apparatus and intercellular gaps of the central nervous tissue. The possibility of the virus spread both throughout the cytoplasm of nerve cells and their processes and the extracellular spaces of the brain was confirmed. Localization of virions within neurons, synapses and myelinated fibers of the spinal cord after intracerebral inoculation suggests that virus spread in the CNS can occur through the CNS parenchyma and also through the nervous conduction pathways. The possible mechanisms of virus dissemination in the CNS of albino mice with experimental Powassan virus encephalomyelitis are discussed.


Subject(s)
Central Nervous System/microbiology , Encephalitis Viruses, Tick-Borne/pathogenicity , Encephalitis, Tick-Borne/microbiology , Neurons/microbiology , Animals , Axons/microbiology , Brain/microbiology , Dendrites/microbiology , Inclusion Bodies, Viral , Mice , Neural Pathways/microbiology , Spinal Cord/microbiology , Virion/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL