Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
1.
Curr Protoc ; 3(7): e841, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37486157

ABSTRACT

Our ability to understand the function of the nervous system is dependent upon defining the connections of its constituent neurons. Development of methods to define connections within neural networks has always been a growth industry in the neurosciences. Transneuronal spread of neurotropic viruses currently represents the best means of defining synaptic connections within neural networks. The method exploits the ability of viruses to invade neurons, replicate, and spread through the intimate synaptic connections that enable communication among neurons. Since the method was first introduced in the 1970s, it has benefited from an increased understanding of the virus life cycle, the function of viral genomes, and the ability to manipulate the viral genome in support of directional spread of virus and the expression of transgenes. In this article, we review these advances in viral tracing technology and the ways in which they may be applied for functional dissection of neural networks. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Retrograde infection of CNS circuits by peripheral injection of virus Basic Protocol 2: Transneuronal analysis by intracerebral injection Alternate Protocol 1: Transneuronal analysis with multiple recombinant strains Alternate Protocol 2: Conditional replication and spread of PRV Alternate Protocol 3: Conditional reporters of PRV infection and spread Alternate Protocol 4: Reporters of neural activity in polysynaptic circuits Support Protocol 1: Growing and titering a PRV viral stock Support Protocol 2: Immunohistochemical processing and detection Support Protocol 3: Dual-immunofluorescence localization.


Subject(s)
Herpesvirus 1, Suid , Animals , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/metabolism , Neurons/metabolism
2.
J Neurosci Methods ; 308: 228-239, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30098326

ABSTRACT

BACKGROUND: Viral transneuronal tracing has become a well established technology used to define the synaptic architecture of polysynaptic neural networks. NEW METHOD: In this report we define the neuroinvasive profile and reporter expression of a new recombinant of the Bartha strain of pseudorabies virus (PRV). The new recombinant, PRV-290, expresses the mTurquoise2 fluorophor and is designed to complement other isogenic recombinants of Bartha that express different reporters of infection. Results & Comparison with Existing Methods: PRV-290 was injected either alone or in combination with isogenic recombinants of PRV that express enhanced green fluorescent protein (EGFP; PRV-152) or monomeric red fluorescent protein (mRFP; PRV-614). Circuits previously defined using PRV-152 and PRV-614 were used for the analysis. The data demonstrate that PRV-290 is a retrograde transneuronal tracer with temporal kinetics similar to those of its isogenic recombinants. Stable expression of the diffusible mTurquoise2 reporter filled infected neurons, with the extent and intensity of labeling increasing with advancing post inoculation survival. In multiple injection experiments, PRV-290 established productive infections in neurons also replicating PRV-152 and/or PRV-614. This novel demonstration of three recombinants infecting individual neurons represents an important advance in the technology. CONCLUSION: Collectively, these data demonstrate that PRV-290 is a valuable addition to the viral tracer toolbox for transneuronal tracing of neural circuitry.


Subject(s)
Brain/cytology , Brain/virology , Herpesvirus 1, Suid/physiology , Neuroanatomical Tract-Tracing Techniques/methods , Neurons/virology , Animals , Cell Line , Genetic Vectors , Male , Neural Pathways/cytology , Neural Pathways/virology , Neuronal Tract-Tracers , Neurons/cytology , Rats, Sprague-Dawley , Viscera/cytology , Viscera/virology
3.
J Comp Neurol ; 526(14): 2149-2164, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30019398

ABSTRACT

Glutamatergic neurons that express pre-proglucagon (PPG) and are immunopositive (+) for glucagon-like peptide-1 (i.e., GLP-1+ neurons) are located within the caudal nucleus of the solitary tract (cNTS) and medullary reticular formation in rats and mice. GLP-1 neurons give rise to an extensive central network in which GLP-1 receptor (GLP-1R) signaling suppresses food intake, attenuates rewarding, increases avoidance, and stimulates stress responses, partly via GLP-1R signaling within the cNTS. In mice, noradrenergic (A2) cNTS neurons express GLP-1R, whereas PPG neurons do not. In this study, confocal microscopy in rats confirmed that prolactin-releasing peptide (PrRP)+ A2 neurons are closely apposed by GLP-1+ axonal varicosities. Surprisingly, GLP-1+ appositions were also observed on dendrites of PPG/GLP-1+ neurons in both species, and electron microscopy in rats revealed that GLP-1+ boutons form asymmetric synaptic contacts with GLP-1+ dendrites. However, RNAscope confirmed that rat GLP-1 neurons do not express GLP-1R mRNA. Similarly, Ca2+ imaging of somatic and dendritic responses in mouse ex vivo slices confirmed that PPG neurons do not respond directly to GLP-1, and a mouse crossbreeding strategy revealed that <1% of PPG neurons co-express GLP-1R. Collectively, these data suggest that GLP-1R signaling pathways modulate the activity of PrRP+ A2 neurons, and also reveal a local "feed-forward" synaptic network among GLP-1 neurons that apparently does not use GLP-1R signaling. This local GLP-1 network may instead use glutamatergic signaling to facilitate dynamic and potentially selective recruitment of GLP-1 neural populations that shape behavioral and physiological responses to internal and external challenges.


Subject(s)
Glucagon-Like Peptide 1/physiology , Nerve Net/physiology , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Synapses/physiology , Animals , Female , Glucagon-Like Peptide-1 Receptor/biosynthesis , Glucagon-Like Peptide-1 Receptor/genetics , Glutamate Decarboxylase , Male , Mice , Mice, Transgenic , Nerve Net/cytology , Proglucagon/metabolism , Prolactin-Releasing Hormone/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Solitary Nucleus/ultrastructure , Synapses/ultrastructure
5.
J Virol ; 90(1): 379-91, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26491149

ABSTRACT

UNLABELLED: Varicella-zoster virus (VZV) is a highly neurotropic virus that can cause infections in both the peripheral nervous system and the central nervous system. Several studies of VZV reactivation in the peripheral nervous system (herpes zoster) have been published, while exceedingly few investigations have been carried out in a human brain. Notably, there is no animal model for VZV infection of the central nervous system. In this report, we characterized the cellular environment in the temporal lobe of a human subject who recovered from focal VZV encephalitis. The approach included not only VZV DNA/RNA analyses but also a delineation of infected cell types (neurons, microglia, oligodendrocytes, and astrocytes). The average VZV genome copy number per cell was 5. Several VZV regulatory and structural gene transcripts and products were detected. When colocalization studies were performed to determine which cell types harbored the viral proteins, the majority of infected cells were astrocytes, including aggregates of astrocytes. Evidence of syncytium formation within the aggregates included the continuity of cytoplasm positive for the VZV glycoprotein H (gH) fusion-complex protein within a cellular profile with as many as 80 distinct nuclei. As with other causes of brain injury, these results suggested that astrocytes likely formed a defensive perimeter around foci of VZV infection (astrogliosis). Because of the rarity of brain samples from living humans with VZV encephalitis, we compared our VZV results with those found in a rat encephalitis model following infection with the closely related pseudorabies virus and observed similar perimeters of gliosis. IMPORTANCE: Investigations of VZV-infected human brain from living immunocompetent human subjects are exceedingly rare. Therefore, much of our knowledge of VZV neuropathogenesis is gained from studies of VZV-infected brains obtained at autopsy from immunocompromised patients. These are not optimal samples with which to investigate a response by a human host to VZV infection. In this report, we examined both flash-frozen and paraffin-embedded formalin-fixed brain tissue of an otherwise healthy young male with focal VZV encephalitis, most likely acquired from VZV reactivation in the trigeminal ganglion. Of note, the cellular response to VZV infection mimicked the response to other causes of trauma to the brain, namely, an ingress of astrocytes and astrogliosis around an infectious focus. Many of the astrocytes themselves were infected; astrocytes aggregated in clusters. We postulate that astrogliosis represents a successful defense mechanism by an immunocompetent human host to eliminate VZV reactivation within neurons.


Subject(s)
Astrocytes/immunology , Encephalitis, Varicella Zoster/pathology , Gliosis/pathology , Herpesvirus 3, Human/immunology , Animals , Astrocytes/virology , Disease Models, Animal , Encephalitis, Varicella Zoster/immunology , Encephalitis, Varicella Zoster/virology , Giant Cells/pathology , Giant Cells/virology , Gliosis/immunology , Herpesvirus 1, Suid , Humans , Male , Pseudorabies/immunology , Pseudorabies/pathology , Pseudorabies/virology , Rats, Sprague-Dawley , Temporal Lobe/pathology , Temporal Lobe/virology
6.
Brain Struct Funct ; 220(3): 1395-420, 2015.
Article in English | MEDLINE | ID: mdl-24585022

ABSTRACT

The use of viruses as transneuronal tracers has become an increasingly powerful technique for defining the synaptic organization of neural networks. Although a number of recombinant alpha herpesviruses are known to spread selectively in the retrograde direction through neural circuits only one strain, the H129 strain of herpes simplex virus type 1, is reported to selectively spread in the anterograde direction. However, it is unclear from the literature whether there is an absolute block or an attenuation of retrograde spread of H129. Here, we demonstrate efficient anterograde spread, and temporally delayed retrograde spread, of H129 and three novel recombinants. In vitro studies revealed no differences in anterograde and retrograde spread of parental H129 and its recombinants through superior cervical ganglion neurons. In vivo injections of rat striatum revealed a clear bias of anterograde spread, although evidence of deficient retrograde transport was also present. Evidence of temporally delayed retrograde transneuronal spread of H129 in the retina was observed following injection of the lateral geniculate nucleus. The data also demonstrated that three novel recombinants efficiently express unique fluorescent reporters and have the capacity to infect the same neurons in dual infection paradigms. From these experiments we conclude that H129 and its recombinants not only efficiently infect neurons through anterograde transneuronal passage, but also are capable of temporally delayed retrograde transneuronal spread. In addition, the capacity to produce dual infection of projection targets following anterograde transneuronal passage provides an important addition to viral transneuronal tracing technology.


Subject(s)
Brain/cytology , Brain/virology , Herpesvirus 1, Human/physiology , Neuroanatomical Tract-Tracing Techniques/methods , Neurons/cytology , Neurons/virology , Animals , Antigens, Viral , Axonal Transport , Basal Ganglia/cytology , Basal Ganglia/virology , Cells, Cultured , Chlorocebus aethiops , Genes, Reporter , Gliosis/pathology , Gliosis/virology , Humans , Male , Rats , Rats, Sprague-Dawley , Visual Pathways/cytology , Visual Pathways/virology
7.
Curr Protoc Neurosci ; 68: 1.5.1-1.5.39, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24984685

ABSTRACT

Our ability to understand the function of the nervous system is dependent upon defining the connections of its constituent neurons. Development of methods to define connections within neural networks has always been a growth industry in the neurosciences. Transneuronal spread of neurotropic viruses currently represents the best means of defining synaptic connections within neural networks. The method exploits the ability of viruses to invade neurons, replicate, and spread through the intimate synaptic connections that enable communication among neurons. Since the method was first introduced in the 1970s, it has benefited from an increased understanding of the virus life cycle, the function of viral genome, and the ability to manipulate the viral genome in support of directional spread of virus and the expression of transgenes. In this unit, we review these advances in viral tracing technology and the way in which they may be applied for functional dissection of neural networks.


Subject(s)
Brain/cytology , Herpesvirus 1, Suid/chemistry , Nerve Net/chemistry , Nerve Net/cytology , Neurons/chemistry , Animals , Brain/metabolism , Nerve Net/metabolism , Neurons/metabolism , Rodentia
8.
Childs Nerv Syst ; 30(8): 1333-42, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24881033

ABSTRACT

PURPOSE: Pediatric traumatic brain injury (TBI) represents a prominent yet understudied medical condition that can profoundly impact brain development. As the juvenile injured brain matures in the wake of neuropathological cascades during potentially critical periods, circuit alterations may explain neurological consequences, including cognitive deficits. We hypothesize that experimental brain injury in juvenile rats, with behavioral deficits that resolve, will lead to quantifiable structural changes in hippocampal neurons at chronic time points post-injury. METHODS: Controlled cortical impact (CCI), a model of focal TBI with contusion, was used to induce brain injury on post-natal day (PND) 17 juvenile rats. The histological consequence of TBI was quantified in regions of the hippocampus at post-injury day 28 (PID28) on sections stained using a variation of the Golgi-Cox staining method. Individual neuronal morphologies were digitized from the dentate gyrus (DG), CA3, and CA1 regions. RESULTS: Soma area in the ipsilateral injured DG and CA3 regions of the hippocampus increased significantly at PID28 in comparison to controls. In CA1, dendritic length and dendritic branching decreased significantly in comparison to controls and the contralateral hemisphere, without change in soma area. To extend the study, we examined neuronal morphology in rats with CCI at PND7. On PID28 after CCI on PND7 rats, CA1 neurons showed no injury-induced change in morphology, potentially indicating an age-dependent morphological response to injury. CONCLUSIONS: Long-lasting structural alterations in hippocampal neurons of brain-injured PND17 juvenile animals, but not PND7 immature animals, suggest differential plasticity depending on age-at-injury, with potential consequences for later function.


Subject(s)
Brain Injuries/pathology , Hippocampus/pathology , Neurons/pathology , Age Factors , Animals , Animals, Newborn , Cerebral Cortex/pathology , Dendrites/pathology , Dendrites/ultrastructure , Female , Male , Neurons/ultrastructure , Pregnancy , Rats , Rats, Sprague-Dawley , Silver Staining
9.
PLoS One ; 9(4): e93484, 2014.
Article in English | MEDLINE | ID: mdl-24695551

ABSTRACT

Bardet-Biedl syndrome (BBS) is a genetically heterogeneous inherited human disorder displaying a pleotropic phenotype. Many of the symptoms characterized in the human disease have been reproduced in animal models carrying deletions or knock-in mutations of genes causal for the disorder. Thinning of the cerebral cortex, enlargement of the lateral and third ventricles, and structural changes in cilia are among the pathologies documented in these animal models. Ciliopathy is of particular interest in light of recent studies that have implicated primary neuronal cilia (PNC) in neuronal signal transduction. In the present investigation, we tested the hypothesis that areas of the brain responsible for learning and memory formation would differentially exhibit PNC abnormalities in animals carrying a deletion of the Bbs4 gene (Bbs4-/-). Immunohistochemical localization of adenylyl cyclase-III (ACIII), a marker restricted to PNC, revealed dramatic alterations in PNC morphology and a statistically significant reduction in number of immunopositive cilia in the hippocampus and amygdala of Bbs4-/- mice compared to wild type (WT) littermates. Western blot analysis confirmed the decrease of ACIII levels in the hippocampus and amygdala of Bbs4-/- mice, and electron microscopy demonstrated pathological alterations of PNC in the hippocampus and amygdala. Importantly, no neuronal loss was found within the subregions of amygdala and hippocampus sampled in Bbs4-/- mice and there were no statistically significant alterations of ACIII immunopositive cilia in other areas of the brain not known to contribute to the BBS phenotype. Considered with data documenting a role of cilia in signal transduction these findings support the conclusion that alterations in cilia structure or neurochemical phenotypes may contribute to the cognitive deficits observed in the Bbs4-/- mouse mode.


Subject(s)
Amygdala/pathology , Bardet-Biedl Syndrome/pathology , Cilia/pathology , Hippocampus/pathology , Adenylyl Cyclases/metabolism , Amygdala/metabolism , Animals , Bardet-Biedl Syndrome/metabolism , Cilia/metabolism , Disease Models, Animal , Hippocampus/metabolism , Male , Mice , Microtubule-Associated Proteins/metabolism
10.
Neuroscience ; 227: 247-59, 2012 Dec 27.
Article in English | MEDLINE | ID: mdl-23041757

ABSTRACT

C1 catecholamine neurons reside within the rostroventrolateral medulla (RVLM), an area that plays an integral role in blood pressure regulation through reticulospinal projections to sympathetic preganglionic neurons in the thoracic spinal cord. In a previous investigation we mapped the efferent projections of C1 neurons, documenting supraspinal projections to cell groups in the preautonomic network that contribute to the control of cardiovascular function. Light microscopic study also revealed putative local circuit connections within RVLM. In this investigation we tested the hypothesis that RVLM C1 neurons elaborate a local circuit synaptic network that permits communication between C1 neurons giving rise to supraspinal and reticulospinal projections. A replication defective lentivirus vector that expresses enhanced green fluorescent protein (EGFP) under the control of a synthetic dopamine beta hydroxylase (DßH) promoter was used to label C1 neurons and their processes. Confocal fluorescence microscopy demonstrated thin varicose axons immunopositive for EGFP and tyrosine hydroxylase that formed close appositions to C1 somata and dendrites throughout the rostrocaudal extent of the C1 area. Dual-labeled electron microscopic analysis revealed axosomatic, axodendritic and axospinous synaptic contacts with C1 and non-C1 neurons with a distribution recapitulating that observed in the light microscopic analysis. Labeled boutons were large, contained light axoplasm, lucent spherical vesicles, and formed asymmetric synaptic contacts. Collectively these data demonstrate that C1 neurons form a synaptic network within the C1 area that may function to coordinate activity among projection-specific subpopulations of neurons. The data also suggest that the boundaries of RVLM should be defined on the basis of function criteria rather than the C1 phenotype of neurons.


Subject(s)
Catecholamines/metabolism , Medulla Oblongata/cytology , Nerve Net/physiology , Neurons/physiology , Synapses/metabolism , Animals , Brain Mapping , Dopamine beta-Hydroxylase/metabolism , Genetic Vectors/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Microscopy, Immunoelectron , Nerve Net/ultrastructure , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/ultrastructure , Transduction, Genetic , Tyrosine 3-Monooxygenase/metabolism
11.
PLoS One ; 6(6): e21141, 2011.
Article in English | MEDLINE | ID: mdl-21698154

ABSTRACT

Replication and transneuronal transport of pseudorabies virus (PRV) are widely used to define the organization of neural circuits in rodent brain. Here we report a dual infection approach that highlights connections to neurons that collateralize within complex networks. The method combines Cre recombinase (Cre) expression from a PRV recombinant (PRV-267) and Cre-dependent reporter gene expression from a second infecting strain of PRV (PRV-263). PRV-267 expresses both Cre and a monomeric red fluorescent protein (mRFP) fused to viral capsid protein VP26 (VP26-mRFP) that accumulates in infected cell nuclei. PRV-263 carries a Brainbow cassette and expresses a red (dTomato) reporter that fills the cytoplasm. However, in the presence of Cre, the dTomato gene is recombined from the cassette, eliminating expression of the red reporter and liberating expression of either yellow (EYFP) or cyan (mCerulean) cytoplasmic reporters. We conducted proof-of-principle experiments using a well-characterized model in which separate injection of recombinant viruses into the left and right kidneys produces infection of neurons in the renal preautonomic network. Neurons dedicated to one kidney expressed the unique reporters characteristic of PRV-263 (cytoplasmic dTomato) or PRV-267 (nuclear VP26-mRFP). Dual infected neurons expressed VP26-mRFP and the cyan or yellow cytoplasmic reporters activated by Cre-mediated recombination of the Brainbow cassette. Differential expression of cyan or yellow reporters in neurons lacking VP26-mRFP provided a unique marker of neurons synaptically connected to dual infected neurons, a synaptic relationship that cannot be distinguished using other dual infection tracing approaches. These data demonstrate Cre-enabled conditional reporter expression in polysynaptic circuits that permits the identification of collateralized neurons and their presynaptic partners.


Subject(s)
Herpesvirus 1, Suid/physiology , Neurons/cytology , Pseudorabies/physiopathology , Animals , Base Sequence , Genes, Reporter , Genome, Viral , Herpesvirus 1, Suid/genetics , Male , Microscopy, Fluorescence , Neurons/virology , Open Reading Frames , Rats , Rats, Sprague-Dawley
12.
Proc Natl Acad Sci U S A ; 108(8): 3377-82, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21292985

ABSTRACT

Transneuronal transport of neurotropic viruses is widely used to define the organization of neural circuitry in the mature and developing nervous system. However, interconnectivity within complex circuits limits the ability of viral tracing to define connections specifically linked to a subpopulation of neurons within a network. Here we demonstrate a unique viral tracing technology that highlights connections to defined populations of neurons within a larger labeled network. This technology was accomplished by constructing a replication-competent strain of pseudorabies virus (PRV-263) that changes the profile of fluorescent reporter expression in the presence of Cre recombinase (Cre). The viral genome carries a Brainbow cassette that expresses a default red reporter in infected cells. However, in the presence of Cre, the red reporter gene is excised from the genome and expression of yellow or cyan reporters is enabled. We used PRV-263 in combination with a unique lentivirus vector that produces Cre expression in catecholamine neurons. Projection-specific infection of central circuits containing these Cre-expressing catecholamine neurons with PRV-263 resulted in Cre-mediated recombination of the PRV-263 genome and conditional expression of cyan/yellow reporters. Replication and transneuronal transport of recombined virus produced conditional reporter expression in neurons synaptically linked to the Cre-expressing catecholamine neurons. This unique technology highlights connections specific to phenotypically defined neurons within larger networks infected by retrograde transneuronal transport of virus from a defined projection target. The availability of other technologies that restrict Cre expression to defined populations of neurons indicates that this approach can be widely applied across functionally defined systems.


Subject(s)
Herpesvirus 1, Suid/genetics , Microdissection/methods , Nerve Net/anatomy & histology , Technology/methods , Biological Transport , Catecholamines , Fluorescent Dyes , Genes, Reporter , Integrases , Nerve Net/cytology , Neurons/chemistry , Neurons/virology
13.
J Comp Neurol ; 518(12): 2202-20, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20437524

ABSTRACT

Phox2a is a transcription factor that plays an essential role, with Phox2b, in the specification of the adrenergic and noradrenergic phenotype in developing brain. Localization of Phox2a in developing brainstem has demonstrated a high degree of correspondence between neurons expressing the transcription factor and those involved in the regulation of autonomic function. Although it is well established that the paralogous gene product Phox2b is widely expressed in adult brain, no study has mapped the distribution of Phox2a in the adult. The data reported here address that void. A well-characterized rabbit polyclonal antiserum was used for immunohistochemical localization of the transcription factor in adult rats. Sections through the rostrocaudal extent of brain were processed for dual immunocytochemical localization of Phox2a and catecholamine enzymes. Adjacent sections were used for dual localization of Phox2a and NADPH diaphorase, a marker of nitric oxide-containing neurons. The data demonstrate that Phox2a is present in all brainstem catecholamine neurons, in circumscribed populations of NADPH(+) neurons, and in a subset of neurons that influences sympathetic and parasympathetic outflow. In addition, Phox2a(+) neurons were observed within diencephalic and brainstem nuclei that regulate behavioral state. Considered with data demonstrating that Phox2a is part of the transcriptional complex that drives expression of dopamine-beta-hydroxylase and can also up-regulate expression of other genes, the data support the conclusion that Phox2a plays an important role in brainstem catecholamine neurotransmission and in the regulation of adaptive homeostatic functions in the adult nervous system.


Subject(s)
Brain/cytology , Brain/metabolism , Homeodomain Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Aging , Animals , Brain Stem/cytology , Brain Stem/metabolism , Catecholamines/metabolism , Cell Count , Diencephalon/cytology , Diencephalon/metabolism , Dopamine beta-Hydroxylase/metabolism , Female , Immunohistochemistry , Male , NADPH Dehydrogenase/metabolism , Phenotype , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
14.
J Neurosci ; 30(8): 2844-55, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181582

ABSTRACT

Hippocampal mossy fiber (MF) synapses on area CA3 lacunosum-moleculare (L-M) interneurons are capable of undergoing a Hebbian form of NMDA receptor (NMDAR)-independent long-term potentiation (LTP) induced by the same type of high-frequency stimulation (HFS) that induces LTP at MF synapses on pyramidal cells. LTP of MF input to L-M interneurons occurs only at synapses containing mostly calcium-impermeable (CI)-AMPA receptors (AMPARs). Here, we demonstrate that HFS-induced LTP at these MF-interneuron synapses requires postsynaptic activation of protein kinase A (PKA) and protein kinase C (PKC). Brief extracellular stimulation of PKA with forskolin (FSK) alone or in combination with 1-Methyl-3-isobutylxanthine (IBMX) induced a long-lasting synaptic enhancement at MF synapses predominantly containing CI-AMPARs. However, the FSK/IBMX-induced potentiation in cells loaded with the specific PKA inhibitor peptide PKI(6-22) failed to be maintained. Consistent with these data, delivery of HFS to MFs synapsing onto L-M interneurons loaded with PKI(6-22) induced posttetanic potentiation (PTP) but not LTP. Hippocampal sections stained for the catalytic subunit of PKA revealed abundant immunoreactivity in interneurons located in strata radiatum and L-M of area CA3. We also found that extracellular activation of PKC with phorbol 12,13-diacetate induced a pharmacological potentiation of the isolated CI-AMPAR component of the MF EPSP. However, HFS delivered to MF synapses on cells loaded with the PKC inhibitor chelerythrine exhibited PTP followed by a significant depression. Together, our data indicate that MF LTP in L-M interneurons at synapses containing primarily CI-AMPARs requires some of the same signaling cascades as does LTP of glutamatergic input to CA3 or CA1 pyramidal cells.


Subject(s)
CA3 Region, Hippocampal/enzymology , Interneurons/enzymology , Long-Term Potentiation/physiology , Mossy Fibers, Hippocampal/enzymology , Protein Kinases/metabolism , Synaptic Transmission/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Benzophenanthridines/pharmacology , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , Catalytic Domain/drug effects , Catalytic Domain/physiology , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Glutamic Acid/metabolism , Interneurons/cytology , Interneurons/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Long-Term Potentiation/drug effects , Male , Mossy Fibers, Hippocampal/drug effects , Mossy Fibers, Hippocampal/ultrastructure , Organ Culture Techniques , Peptide Fragments/pharmacology , Phorbol Esters/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Protein Kinases/drug effects , Rats , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptic Membranes/drug effects , Synaptic Membranes/enzymology , Synaptic Transmission/drug effects
15.
J Comp Neurol ; 515(6): 677-95, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19496174

ABSTRACT

The morphological and electrophysiological diversity of inhibitory cells in hippocampal area CA3 may underlie specific computational roles and is not yet fully elucidated. In particular, interneurons with somata in strata radiatum (R) and lacunosum-moleculare (L-M) receive converging stimulation from the dentate gyrus and entorhinal cortex as well as within CA3. Although these cells express different forms of synaptic plasticity, their axonal trees and connectivity are still largely unknown. We investigated the branching and spatial patterns, plus the membrane and synaptic properties, of rat CA3b R and L-M interneurons digitally reconstructed after intracellular labeling. We found considerable variability within but no difference between the two layers, and no correlation between morphological and biophysical properties. Nevertheless, two cell types were identified based on the number of dendritic bifurcations, with significantly different anatomical and electrophysiological features. Axons generally branched an order of magnitude more than dendrites. However, interneurons on both sides of the R/L-M boundary revealed surprisingly modular axodendritic arborizations with consistently uniform local branch geometry. Both axons and dendrites followed a lamellar organization, and axons displayed a spatial preference toward the fissure. Moreover, only a small fraction of the axonal arbor extended to the outer portion of the invaded volume, and tended to return toward the proximal region. In contrast, dendritic trees demonstrated more limited but isotropic volume occupancy. These results suggest a role of predominantly local feedforward and lateral inhibitory control for both R and L-M interneurons. Such a role may be essential to balance the extensive recurrent excitation of area CA3 underlying hippocampal autoassociative memory function.


Subject(s)
Hippocampus/cytology , Interneurons , Animals , Axons/ultrastructure , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials , Interneurons/classification , Interneurons/cytology , Interneurons/metabolism , Male , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism
16.
J Virol ; 83(3): 1173-83, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19019952

ABSTRACT

The attenuated pseudorabies virus (PRV) strain Bartha contains several characterized mutations that affect its virulence and ability to spread through neural circuits. This strain contains a small genomic deletion that abrogates anterograde spread and is widely used as a retrograde-restricted neural circuit tracer. Previous studies showed that the retrograde-directed spread of PRV Bartha is slower than that of wild-type PRV. We used compartmented neuronal cultures to characterize the retrograde defect and identify the genetic basis of the phenotype. PRV Bartha is not impaired in retrograde axonal transport, but transneuronal spread among neurons is diminished. Repair of the U(L)21 locus with wild-type sequence restored efficient transneuronal spread both in vitro and in vivo. It is likely that mutations in the Bartha U(L)21 gene confer defects that affect infectious particle production, causing a delay in spread to presynaptic neurons and amplification of infection. These events manifest as slower kinetics of retrograde viral spread in a neural circuit.


Subject(s)
Capsid Proteins/genetics , Herpesvirus 1, Suid/genetics , Neurons/virology , Animals , Fluorescent Antibody Technique , Herpesvirus 1, Suid/physiology , Male , Rats , Rats, Sprague-Dawley
17.
J Physiol ; 586(11): 2695-712, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18388134

ABSTRACT

We performed whole-cell recordings from CA3 s. radiatum (R) and s. lacunosum-moleculare (L-M) interneurons in hippocampal slices to examine the temporal aspects of summation of converging perforant path (PP) and mossy fibre (MF) inputs. PP EPSPs were evoked from the s. lacunosum-moleculare in area CA1. MF EPSPs were evoked from the medial extent of the suprapyramidal blade of the dentate gyrus. Summation was strongly supralinear when examining PP EPSP with MF EPSP in a heterosynaptic pair at the 10 ms ISI, and linear to sublinear at longer ISIs. This pattern of nonlinearities suggests that R and L-M interneurons act as coincidence detectors for input from PP and MF. Summation at all ISIs was linear in voltage clamp mode demonstrating that nonlinearities were generated by postsynaptic voltage-dependent conductances. Supralinearity was not detected when the first EPSP in the pair was replaced by a simulated EPSP injected into the soma, suggesting that the conductances underlying the EPSP boosting were located in distal dendrites. Supralinearity was selectively eliminated with either Ni2+ (30 microm), mibefradil (10 microm) or nimodipine (15 microm), but was unaffected by QX-314. This pharmacological profile indicates that supralinearity is due to recruitment of dendritic T-type Ca2+channels by the first subthreshold EPSP in the pair. Results with the hyperpolarization-activated (Ih) channel blocker ZD 7288 (50 microm) revealed that Ih restricted the time course of supralinearity for coincidently summed EPSPs, and promoted linear to sublinear summation for asynchronous EPSPs. We conclude that coincidence detection results from the counterbalanced activation of T-type Ca2+ channels and inactivation of Ih.


Subject(s)
Action Potentials/physiology , Evoked Potentials/physiology , Hippocampus/physiology , Mossy Fibers, Hippocampal/physiology , Perforant Pathway/physiology , Animals , Male , Rats , Rats, Sprague-Dawley
18.
Neuroinformatics ; 6(1): 63-7, 2008.
Article in English | MEDLINE | ID: mdl-18350259

ABSTRACT

The freeware Java tool Point Analysis in Java (PAJ), created to perform 3D point analysis, was tested in an independent laboratory setting. The input data consisted of images of the hippocampal perforant pathway from serial immunocytochemical localizations of the rat brain in multiple views at different resolutions. The low magnification set (x2 objective) comprised the entire perforant pathway, while the high magnification set (x100 objective) allowed the identification of individual fibers. A preliminary stereological study revealed a striking linear relationship between the fiber count at high magnification and the optical density at low magnification. PAJ enabled fast analysis for down-sampled data sets and a friendly interface with automated plot drawings. Noted strengths included the multi-platform support as well as the free availability of the source code, conducive to a broad user base and maximum flexibility for ad hoc requirements. PAJ has great potential to extend its usability by (a) improving its graphical user interface, (b) increasing its input size limit, (c) improving response time for large data sets, and (d) potentially being integrated with other Java graphical tools such as ImageJ.


Subject(s)
Axons/ultrastructure , Computer Simulation/standards , Image Cytometry/methods , Neuroanatomy/methods , Perforant Pathway/cytology , Software/standards , Access to Information , Algorithms , Animals , Axons/physiology , Brain Mapping/methods , Computer Simulation/trends , Image Cytometry/trends , Internet/trends , Neuroanatomy/trends , Perforant Pathway/physiology , Rats , Software/trends , Software Validation , Time Factors
19.
Neuroscience ; 146(3): 890-900, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17391860

ABSTRACT

Bleomycin hydrolase (BLMH) is a multifaceted neutral cysteine protease with a suggested role in antigen presentation, homocysteine-thiolactone metabolism, and Alzheimer's disease pathogenesis. Deletion of the protease in mice results in increased neonatal mortality and dermatopathology. Immunohistochemical and behavioral studies of BLMH knockout mice were undertaken to further evaluate the role of the protease in the brain. No gross abnormalities in the CNS were observed upon preliminary histological examination of B6.129Blmhtm1Geh/J null animals. However, glial fibrillary acid protein immunohistochemistry revealed a global reactive astrogliosis in the aged null animals, indicative of undefined brain pathology. The role of BLMH in the brain was further explored by characterizing the behavioral phenotype of hybrid [129S6-Blmhtm1Geh/JxB6.129 Blmhtm1Geh/J]F1 null and littermate controls using multiple behavioral paradigms. In the water maze, deletion of BLMH resulted in poorer performance during water maze probe trials without detectable effect of the mutation on sensorimotor function. In addition, no age-dependent decline in discriminative performance on probe trials was observed in null animals. These data suggest a physiological non-redundant function for BLMH in the CNS.


Subject(s)
Behavior, Animal/physiology , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/physiology , Gliosis/genetics , Gliosis/psychology , Animals , Brain/enzymology , Brain/physiology , Conditioning, Operant/physiology , Cues , DNA/genetics , Exploratory Behavior/physiology , Fear/psychology , Female , Genotype , Hippocampus/physiology , Immunohistochemistry , Light , Male , Maze Learning/physiology , Mice , Mice, Knockout , Postural Balance/physiology
20.
J Comp Neurol ; 499(5): 840-59, 2006 Dec 10.
Article in English | MEDLINE | ID: mdl-17048222

ABSTRACT

A replication-defective lentivirus vector that expresses enhanced green fluorescent protein (EGFP) under the control of a synthetic dopamine-beta-hydroxylase (DbetaH) promoter was used to define efferent projections of C1 catecholamine neurons in rat rostral ventrolateral medulla (RVLM). EGFP expression was restricted to C1 neurons and filled their somatodendritic compartments and efferent axons 7-28 days after vector injection. This included the descending projections to thoracic spinal cord and a network in brainstem, midbrain, and diencephalon. In caudal brainstem, restricted terminal fields were present in the dorsal motor vagal complex, A1, raphe pallidus and obscurus, and marginal layer of ventrolateral medulla. Innervation of raphe nuclei was most dense at the level of RVLM, but rostral levels of pallidus were devoid of innervation. A sparse commissural projection to contralateral RVLM was observed, and pericellular arbors were present in the dorsal reticular formation among the projection pathway of catecholamine axons. Rostral brainstem contained a dense innervation of locus coeruleus and the nucleus subcoeruleus. A restricted innervation of the ventrolateral column of the periaqueductal gray distinguished the midbrain. Forebrain labeling was restricted to the diencephalon, where distinctive terminal fields were observed in the paraventricular thalamic nucleus; the lateral hypothalamic area; and the paraventricular, dorsomedial, supraoptic, and median preoptic nuclei of hypothalamus. Projection fibers also coursed through the tuberal hypothalamus into the median eminence. Collectively, these data demonstrate that RVLM C1 neurons modulate the activity of other central cell groups known to participate in the regulation of cardiovascular and autonomic function.


Subject(s)
Cardiovascular Physiological Phenomena , Catecholamines/metabolism , Efferent Pathways/anatomy & histology , Medulla Oblongata/anatomy & histology , Neurons/chemistry , Neurons/cytology , Animals , Dopamine beta-Hydroxylase/genetics , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Male , Neurons/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...