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










Publication year range
1.
eNeuro ; 10(12)2023 Dec.
Article in English | MEDLINE | ID: mdl-37989590

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is a polymodal cation channel that is activated by electrophilic irritants, oxidative stress, cold temperature, and GPCR signaling. TRPA1 expression has been primarily identified in subsets of nociceptive sensory afferents and is considered a target for future analgesics. Nevertheless, TRPA1 has been implicated in other cell types including keratinocytes, epithelium, enterochromaffin cells, endothelium, astrocytes, and CNS neurons. Here, we developed a knock-in mouse that expresses the recombinase FlpO in TRPA1-expressing cells. We crossed the TRPA1Flp mouse with the R26ai65f mouse that expresses tdTomato in a Flp-sensitive manner. We found tdTomato expression correlated well with TRPA1 mRNA expression and sensitivity to TRPA1 agonists in subsets of TRPV1 (transient receptor potential vanilloid receptor type 1)-expressing neurons in the vagal ganglia and dorsal root ganglia (DRGs), although tdTomato expression efficiency was limited in DRG. We observed tdTomato-expressing afferent fibers centrally (in the medulla and spinal cord) and peripherally in the esophagus, gut, airways, bladder, and skin. Furthermore, chemogenetic activation of TRPA1-expressing nerves in the paw evoked flinching behavior. tdTomato expression was very limited in other cell types. We found tdTomato in subepithelial cells in the gut mucosa but not in enterochromaffin cells. tdTomato was also observed in supporting cells within the cochlea, but not in hair cells. Lastly, tdTomato was occasionally observed in neurons in the somatomotor cortex and the piriform area, but not in astrocytes or vascular endothelium. Thus, this novel mouse strain may be useful for mapping and manipulating TRPA1-expressing cells and deciphering the role of TRPA1 in physiological and pathophysiological processes.


Subject(s)
Transient Receptor Potential Channels , Animals , Mice , Ganglia, Spinal/metabolism , Gene Expression , Sensory Receptor Cells/metabolism , Skin , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/metabolism
2.
eNeuro ; 9(2)2022.
Article in English | MEDLINE | ID: mdl-35365503

ABSTRACT

The airways are densely innervated by sensory afferent nerves, whose activation regulates respiration and triggers defensive reflexes (e.g., cough, bronchospasm). Airway innervation is heterogeneous, and distinct afferent subsets have distinct functional responses. However, little is known of the innervation patterns of subsets within the lung. A neuroanatomical map is critical for understanding afferent activation under physiological and pathophysiological conditions. Here, we quantified the innervation of the mouse lung by vagal and dorsal root ganglion (DRG) sensory subsets defined by the expression of Pirt (all afferents), 5HT3 (vagal nodose afferents), Tac1 (tachykinergic afferents), and transient receptor potential vanilloid 1 channel (TRPV1; defensive/nociceptive afferents) using Cre-mediated reporter expression. We found that vagal afferents innervate almost all conducting airways and project into the alveolar region, whereas DRG afferents only innervate large airways. Of the two vagal ganglia, only nodose afferents project into the alveolar region, but both nodose and jugular afferents innervate conducting airways throughout the lung. Many afferents that project into the alveolar region express TRPV1. Few DRG afferents expressed TRPV1. Approximately 25% of blood vessels were innervated by vagal afferents (many were Tac1+). Approximately 10% of blood vessels had DRG afferents (some were Tac1+), but this was restricted to large vessels. Lastly, innervation of neuroepithelial bodies (NEBs) correlated with the cell number within the bodies. In conclusion, functionally distinct sensory subsets have distinct innervation patterns within the conducting airways, alveoli and blood vessels. Physiologic (e.g., stretch) and pathophysiological (e.g., inflammation, edema) stimuli likely vary throughout these regions. Our data provide a neuroanatomical basis for understanding afferent responses in vivo.


Subject(s)
Ganglia, Spinal , Vagus Nerve , Afferent Pathways , Animals , Lung/innervation , Lung/metabolism , Mice , Neurons , Neurons, Afferent/physiology , Nodose Ganglion , Vagus Nerve/metabolism
3.
Am J Physiol Regul Integr Comp Physiol ; 321(5): R672-R686, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34523364

ABSTRACT

Action potentials depend on voltage-gated sodium channels (NaV1s), which have nine α subtypes. NaV1 inhibition is a target for pathologies involving excitable cells such as pain. However, because NaV1 subtypes are widely expressed, inhibitors may inhibit regulatory sensory systems. Here, we investigated specific NaV1s and their inhibition in mouse esophageal mechanoreceptors-non-nociceptive vagal sensory afferents that are stimulated by low threshold mechanical distension, which regulate esophageal motility. Using single fiber electrophysiology, we found mechanoreceptor responses to esophageal distension were abolished by tetrodotoxin. Single-cell RT-PCR revealed that esophageal-labeled TRPV1-negative vagal neurons expressed multiple tetrodotoxin-sensitive NaV1s: NaV1.7 (almost all neurons) and NaV1.1, NaV1.2, and NaV1.6 (in ∼50% of neurons). Inhibition of NaV1.7, using PF-05089771, had a small inhibitory effect on mechanoreceptor responses to distension. Inhibition of NaV1.1 and NaV1.6, using ICA-121341, had a similar small inhibitory effect. The combination of PF-05089771 and ICA-121341 inhibited but did not eliminate mechanoreceptor responses. Inhibition of NaV1.2, NaV1.6, and NaV1.7 using LSN-3049227 inhibited but did not eliminate mechanoreceptor responses. Thus, all four tetrodotoxin-sensitive NaV1s contribute to action potential initiation from esophageal mechanoreceptors terminals. This is different to those NaV1s necessary for vagal action potential conduction, as demonstrated using GCaMP6s imaging of esophageal vagal neurons during electrical stimulation. Tetrodotoxin-sensitive conduction was abolished in many esophageal neurons by PF-05089771 alone, indicating a critical role of NaV1.7. In summary, multiple NaV1 subtypes contribute to electrical signaling in esophageal mechanoreceptors. Thus, inhibition of individual NaV1s would likely have minimal effect on afferent regulation of esophageal motility.


Subject(s)
Action Potentials , Esophagus/innervation , Mechanoreceptors/metabolism , Mechanotransduction, Cellular , Vagus Nerve/metabolism , Voltage-Gated Sodium Channels/metabolism , Action Potentials/drug effects , Animals , Gastrointestinal Motility , Mechanoreceptors/drug effects , Mechanotransduction, Cellular/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Sodium Channel Blockers/pharmacology , Stress, Mechanical , Tetrodotoxin/pharmacology , Time Factors , Vagus Nerve/drug effects , Voltage-Gated Sodium Channels/drug effects , Voltage-Gated Sodium Channels/genetics
4.
eNeuro ; 7(4)2020.
Article in English | MEDLINE | ID: mdl-32669344

ABSTRACT

The ATP-sensitive P2X2 ionotropic receptor plays a critical role in a number of signal processes including taste and hearing, carotid body detection of hypoxia, the exercise pressor reflex and sensory transduction of mechanical stimuli in the airways and bladder. Elucidation of the role of P2X2 has been hindered by the lack of selective tools. In particular, detection of P2X2 using established pharmacological and biochemical techniques yields dramatically different expression patterns, particularly in the peripheral and central nervous systems. Here, we have developed a knock-in P2X2-cre mouse, which we crossed with a cre-sensitive tdTomato reporter mouse to determine P2X2 expression. P2X2 was found in more than 80% of nodose vagal afferent neurons, but not in jugular vagal afferent neurons. Reporter expression correlated in vagal neurons with sensitivity to α,ß methylene ATP (αßmATP). P2X2 was expressed in 75% of petrosal afferents, but only 12% and 4% of dorsal root ganglia (DRG) and trigeminal afferents, respectively. P2X2 expression was limited to very few cell types systemically. Together with the central terminals of P2X2-expressing afferents, reporter expression in the CNS was mainly found in brainstem neurons projecting mossy fibers to the cerebellum, with little expression in the hippocampus or cortex. The structure of peripheral terminals of P2X2-expressing afferents was demonstrated in the tongue (taste buds), carotid body, trachea and esophagus. P2X2 was observed in hair cells and support cells in the cochlear, but not in spiral afferent neurons. This mouse strain provides a novel approach to the identification and manipulation of P2X2-expressing cell types.


Subject(s)
Neurons, Afferent , Receptors, Purinergic P2 , Adenosine Triphosphate , Animals , Ganglia, Spinal , Mice , Neurons , Reflex
5.
Respir Physiol Neurobiol ; 278: 103446, 2020 07.
Article in English | MEDLINE | ID: mdl-32360368

ABSTRACT

Inflammation can increase the excitability of bronchopulmonary C-fibers leading to excessive sensations and reflexes (e.g. wheeze and cough). We have previously shown modulation of peripheral nerve terminal mitochondria by antimycin A causes hyperexcitability in TRPV1-expressing bronchopulmonary C-fibers through the activation of protein kinase C (PKC). Here, we have investigated the PKC isoform responsible for this signaling. We found PKCß1, PKCδ and PKCε were expressed by many vagal neurons, with PKCα and PKCß2 expressed by subsets of vagal neurons. In dissociated vagal neurons, antimycin A caused translocation of PKCα but not the other isoforms, and only in TRPV1-lineage neurons. In bronchopulmonary C-fiber recordings, antimycin A increased the number of action potentials evoked by α,ß-methylene ATP. Selective inhibition of PKCα, PKCß1 and PKCß2 with 50 nM bisindolylmaleimide I prevented the antimycin-induced bronchopulmonary C-fiber hyperexcitability, whereas selective inhibition of only PKCß1 and PKCß2 with 50 nM LY333531 had no effect. We therefore conclude that PKCα is required for antimycin-induced increases in bronchopulmonary C-fiber excitability.


Subject(s)
Antimycin A/pharmacology , Bronchi/innervation , Nerve Fibers, Unmyelinated/drug effects , Neurons/drug effects , Nodose Ganglion/drug effects , Protein Kinase C-alpha/drug effects , Vagus Nerve , Animals , Lung/innervation , Mice , Nerve Fibers, Unmyelinated/metabolism , Neurons/metabolism , Nodose Ganglion/cytology , Nodose Ganglion/metabolism , Protein Isoforms/drug effects , Protein Isoforms/metabolism , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Protein Kinase C-alpha/metabolism , TRPV Cation Channels/metabolism
6.
eNeuro ; 7(2)2020.
Article in English | MEDLINE | ID: mdl-32060036

ABSTRACT

Vagal afferent sensory nerves, originating in jugular and nodose ganglia, are composed of functionally distinct subsets whose activation evokes distinct thoracic and abdominal reflex responses. We used Cre-expressing mouse strains to identify specific vagal afferent populations and map their central projections within the brainstem. We show that Pirt is expressed in virtually all vagal afferents; whereas, 5-HT3 is expressed only in nodose neurons, with little expression in jugular neurons. Transient receptor potential vanilloid 1 (TRPV1), the capsaicin receptor, is expressed in a subset of small nodose and jugular neurons. Tac1, the gene for tachykinins, is expressed predominantly in jugular neurons, some of which also express TRPV1. Vagal fibers project centrally to the nucleus tractus solitarius (nTS), paratrigeminal complex, area postrema, and to a limited extent the dorsal motor nucleus of the vagus. nTS subnuclei preferentially receive projections by specific afferent subsets, with TRPV1+ fibers terminating in medial and dorsal regions predominantly caudal of obex, whereas TRPV1- fibers terminate in ventral and lateral regions throughout the rostral-caudal aspect of the medulla. Many vagal Tac1+ afferents (mostly derived from the jugular ganglion) terminate in the nTS. The paratrigeminal complex was the target of multiple vagal afferent subsets. Importantly, lung-specific TRPV1+ and Tac1+ afferent terminations were restricted to the caudal medial nTS, with no innervation of other medulla regions. In summary, this study identifies the specific medulla regions innervated by vagal afferent subsets. The distinct terminations provide a neuroanatomic substrate for the diverse range of reflexes initiated by vagal afferent activation.


Subject(s)
Nodose Ganglion , Vagus Nerve , Afferent Pathways/metabolism , Animals , Brain Stem/metabolism , Carrier Proteins , Membrane Proteins , Mice , Nodose Ganglion/metabolism , Solitary Nucleus , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Vagus Nerve/metabolism
7.
Brain Res ; 1715: 94-105, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30914247

ABSTRACT

Inflammation causes activation of nociceptive sensory nerves, resulting in debilitating sensations and reflexes. Inflammation also induces mitochondrial dysfunction through multiple mechanisms. Sensory nerve terminals are densely packed with mitochondria, suggesting that mitochondrial signaling may play a role in inflammation-induced nociception. We have previously shown that agents that induce mitochondrial dysfunction, such as antimycin A, activate a subset of nociceptive vagal sensory nerves that express transient receptor potential (TRP) channels ankyrin 1 (A1) and vanilloid 1 (V1). However, the mechanisms underlying these responses are incompletely understood. Here, we studied the contribution of TRPA1, TRPV1 and reactive oxygen species (ROS) to antimycin A-induced vagal sensory nerve activation in dissociated neurons and at the sensory terminals of bronchopulmonary C-fibers. Nociceptive neurons were defined chemically and genetically. Antimycin A-evoked activation of vagal nociceptors in a Fura2 Ca2+ assay correlated with TRPV1 responses compared to TRPA1 responses. Nociceptor activation was dependent on both TRP channels, with TRPV1 predominating in a majority of responding nociceptors and TRPA1 predominating only in nociceptors with the greatest responses. Surprisingly, both TRPA1 and TRPV1 were activated by H2O2 when expressed in HEK293. Nevertheless, targeting ROS had no effect of antimycin A-evoked TRPV1 activation in either HEK293 or vagal neurons. In contrast, targeting ROS inhibited antimycin A-evoked TRPA1 activation in HEK293, vagal neurons and bronchopulmonary C-fibers, and a ROS-insensitive TRPA1 mutant was completely insensitive to antimycin A. We therefore conclude that mitochondrial dysfunction activates vagal nociceptors by ROS-dependent (TRPA1) and ROS-independent (TRPV1) mechanisms.


Subject(s)
Antimycin A/pharmacology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism , Vagus Nerve/metabolism , Action Potentials/drug effects , Animals , Calcium/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Nociceptors/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Transient Receptor Potential Channels/metabolism , Vagus Nerve/cytology , Vagus Nerve/drug effects
8.
BMC Res Notes ; 11(1): 827, 2018 Nov 22.
Article in English | MEDLINE | ID: mdl-30466490

ABSTRACT

OBJECTIVE: Redox-sensitive green fluorescent protein (roGFP) is a genetically-encoded redox-sensitive protein used to detect cellular oxidative stress associated with reactive oxygen species production. Here we replaced the cysteine at position 147 of roGFP1 (variant of roGFP) with selenocysteine in order to increase redox sensitivity of the redox reporter. RESULTS: Expression of roGFP1 selenoprotein (roGFP1-Se147) in HEK293 cells required the presence of a selenocysteine insertion sequence and was augmented by co-expression with SBP2. roGFP1-Se147 demonstrated a similar excitation and emission spectra to roGFP1. Although expression of roGFP1-Se147 was limited, it was sufficient enough to perform live cell imaging to evaluate sensitivity to oxidation and reduction. roGFP1-Se147 exhibited a 100-fold increase in sensitivity to oxidation with H2O2 in comparison to roGFP1 as well as a 20-fold decrease in the EC50 of H2O2. Furthermore, roGFP1-Se147, unlike roGFP1, was able to detect oxidation caused by the mitochondrial electron transport complex III inhibitor antimycin A. Unfortunately roGFP-Se147 exhibited a diminished dynamic range and photoinstability.


Subject(s)
Green Fluorescent Proteins/chemistry , Selenocysteine/chemistry , Antimycin A/chemistry , Cysteine/chemistry , Electron Transport , Glutathione/metabolism , HEK293 Cells , Humans , Hydrogen Peroxide/chemistry , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress , Oxygen/metabolism , Reactive Oxygen Species/metabolism
9.
Mol Pharmacol ; 85(6): 839-48, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24642367

ABSTRACT

Airway sensory nerve excitability is a key determinant of respiratory disease-associated reflexes and sensations such as cough and dyspnea. Inflammatory signaling modulates mitochondrial function and produces reactive oxygen species (ROS). Peripheral terminals of sensory nerves are densely packed with mitochondria; thus, we hypothesized that mitochondrial modulation would alter neuronal excitability. We recorded action potential firing from the terminals of individual bronchopulmonary C-fibers using a mouse ex vivo lung-vagal ganglia preparation. C-fibers were characterized as nociceptors or non-nociceptors based upon conduction velocity and response to transient receptor potential (TRP) channel agonists. Antimycin A (mitochondrial complex III Qi site inhibitor) had no effect on the excitability of non-nociceptors. However, antimycin A increased excitability in nociceptive C-fibers, decreasing the mechanical threshold by 50% and increasing the action potential firing elicited by a P2X2/3 agonist to 270% of control. Antimycin A-induced nociceptor hyperexcitability was independent of TRP ankyrin 1 or TRP vanilloid 1 channels. Blocking mitochondrial ATP production with oligomycin or myxothiazol had no effect on excitability. Antimycin A-induced hyperexcitability was dependent on mitochondrial ROS and was blocked by intracellular antioxidants. ROS are known to activate protein kinase C (PKC). Antimycin A-induced hyperexcitability was inhibited by the PKC inhibitor bisindolylmaleimide (BIM) I, but not by its inactive analog BIM V. In dissociated vagal neurons, antimycin A caused ROS-dependent PKC translocation to the membrane. Finally, H2O2 also induced PKC-dependent nociceptive C-fiber hyperexcitability and PKC translocation. In conclusion, ROS evoked by mitochondrial dysfunction caused nociceptor hyperexcitability via the translocation and activation of PKC.


Subject(s)
Bronchi/innervation , Mitochondria/physiology , Nerve Endings/physiology , Nociceptors/physiology , Protein Kinase C/metabolism , Action Potentials , Animals , Antimycin A/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociceptors/drug effects , Reactive Oxygen Species/metabolism , Sensory Thresholds/drug effects , TRPV Cation Channels/genetics , TRPV Cation Channels/physiology
10.
Brain Res ; 1503: 7-15, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23399679

ABSTRACT

Vagal sensory nerves innervate the majority of visceral organs (e.g., heart, lungs, GI tract, etc) and their activation is critical for defensive and regulatory reflexes. Intracellular Ca(2+) is a key regulator of neuronal excitability and is largely controlled by the Ca(2+) stores of the endoplasmic reticulum. In other cell types store-operated channels (SOC) have been shown to contribute to the homeostatic control of intracellular Ca(2+). Here, using Ca(2+) imaging, we have shown that ER depletion in vagal sensory neurons (using thapsigargin or caffeine) in the absence of extracellular Ca(2+) evoked Ca(2+) influx upon re-introduction of Ca(2+) into the extracellular buffer. This store-operated Ca(2+) entry (SOCE) was observed in approximately 25-40% of vagal neurons, equally distributed among nociceptive and non-nociceptive sensory subtypes. SOCE was blocked by Gd(3+) but not by the Orai channel blocker SKF96365. We found Orai channel mRNA in extracts from whole vagal ganglia, but when using single cell RT-PCR analysis we found only 3 out of 34 neurons expressed Orai channel mRNA, indicating that Orai channel expression in the vagal ganglia was likely derived from non-neuronal cell types. Confocal microscopy of vagal neurons in 3 day cultures demonstrated rich ER tracker fluorescence throughout axonal and neurite structures and ER store depletion (thapsigargin) evoked Ca(2+) transients from these structures. However, no SOCE could be detected in the axonal/neurite structures of vagal neurons. We conclude that SOCE occurs in vagal sensory neuronal cell bodies through non-Orai mechanisms but is absent at nerve terminals.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Sensory Receptor Cells/physiology , Animals , Caffeine/pharmacology , Calcium Channel Blockers/pharmacology , Cells, Cultured , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Imidazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , ORAI1 Protein , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/ultrastructure , TRPA1 Cation Channel , Thapsigargin/pharmacology , Time Factors , Transient Receptor Potential Channels/deficiency , Vagus Nerve/cytology
11.
Mol Pharmacol ; 83(5): 1007-19, 2013 May.
Article in English | MEDLINE | ID: mdl-23444014

ABSTRACT

Mitochondrial dysfunction and subsequent oxidative stress has been reported for a variety of cell types in inflammatory diseases. Given the abundance of mitochondria at the peripheral terminals of sensory nerves and the sensitivity of transient receptor potential (TRP) ankyrin 1 (A1) and TRP vanilloid 1 (V1) to reactive oxygen species (ROS) and their downstream products of lipid peroxidation, we investigated the effect of nerve terminal mitochondrial dysfunction on airway sensory nerve excitability. Here we show that mitochondrial dysfunction evoked by acute treatment with antimycin A (mitochondrial complex III Qi site inhibitor) preferentially activated TRPA1-expressing "nociceptor-like" mouse bronchopulmonary C-fibers. Action potential discharge was reduced by the TRPA1 antagonist HC-030031. Inhibition of TRPV1 further reduced C-fiber activation. In mouse dissociated vagal neurons, antimycin A induced Ca(2+) influx that was significantly reduced by pharmacological inhibition or genetic knockout of either TRPA1 or TRPV1. Inhibition of both TRPA1 and TRPV1 was required to abolish antimycin A-induced Ca(2+) influx in vagal neurons. Using an HEK293 cell expression system, antimycin A induced concentration-dependent activation of both hTRPA1 and hTRPV1 but failed to activate nontransfected cells. Myxothiazol (complex III Qo site inhibitor) inhibited antimycin A-induced TRPA1 activation, as did the reducing agent dithiothreitol. Scavenging of both superoxide and hydrogen peroxide inhibited TRPA1 activation following mitochondrial modulation. In conclusion, we present evidence that acute mitochondrial dysfunction activates airway sensory nerves preferentially via TRPA1 through the actions of mitochondrially-derived ROS. This represents a novel mechanism by which inflammation may be transduced into nociceptive electrical signaling.


Subject(s)
Mitochondria/metabolism , Respiratory System/metabolism , Sensory Receptor Cells/metabolism , Transient Receptor Potential Channels/metabolism , Action Potentials/drug effects , Animals , Antimycin A/pharmacology , Calcium/metabolism , Calcium Channels/metabolism , Cell Line , HEK293 Cells , Humans , Lipid Peroxidation/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Nerve Fibers, Unmyelinated/metabolism , Nerve Tissue Proteins/metabolism , Nociceptors/metabolism , Reactive Oxygen Species/metabolism , Respiratory System/cytology , Respiratory System/drug effects , Sensory Receptor Cells/drug effects , TRPA1 Cation Channel , TRPV Cation Channels/metabolism , Vagus Nerve/drug effects , Vagus Nerve/metabolism
12.
Proc Natl Acad Sci U S A ; 107(12): 5563-8, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20212114

ABSTRACT

Previous research has suggested that human herpesvirus-6 (HHV-6) may integrate into host cell chromosomes and be vertically transmitted in the germ line, but the evidence--primarily fluorescence in situ hybridization (FISH)--is indirect. We sought, first, to definitively test these two hypotheses. Peripheral blood mononuclear cells (PBMCs) were isolated from families in which several members, including at least one parent and child, had unusually high copy numbers of HHV-6 DNA per milliliter of blood. FISH confirmed that HHV-6 DNA colocalized with telomeric regions of one allele on chromosomes 17p13.3, 18q23, and 22q13.3, and that the integration site was identical among members of the same family. Integration of the HHV-6 genome into TTAGGG telomere repeats was confirmed by additional methods and sequencing of the integration site. Partial sequencing of the viral genome identified the same integrated HHV-6A strain within members of families, confirming vertical transmission of the viral genome. We next asked whether HHV-6A infection of naïve cell lines could lead to integration. Following infection of naïve Jjhan and HEK-293 cell lines by HHV-6, the virus integrated into telomeres. Reactivation of integrated HHV-6A virus from individuals' PBMCs as well as cell lines was successfully accomplished by compounds known to induce latent herpesvirus replication. Finally, no circular episomal forms were detected even by PCR. Taken together, the data suggest that HHV-6 is unique among human herpesviruses: it specifically and efficiently integrates into telomeres of chromosomes during latency rather than forming episomes, and the integrated viral genome is capable of producing virions.


Subject(s)
Chromosomes, Human/genetics , Chromosomes, Human/virology , Herpesvirus 6, Human/genetics , Herpesvirus 6, Human/pathogenicity , Telomere/genetics , Telomere/virology , Virus Integration/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Cell Line , Child , DNA, Viral/blood , DNA, Viral/genetics , Female , Gene Dosage , Genome, Viral , Germ Cells/virology , Herpesvirus 6, Human/physiology , Humans , In Situ Hybridization, Fluorescence , In Vitro Techniques , Infectious Disease Transmission, Vertical , Male , Middle Aged , Molecular Sequence Data , Plasmids/blood , Plasmids/genetics , Roseolovirus Infections/genetics , Roseolovirus Infections/transmission , Roseolovirus Infections/virology , Virus Activation , Virus Replication , Young Adult
13.
J Neurosci ; 28(20): 5383-93, 2008 May 14.
Article in English | MEDLINE | ID: mdl-18480294

ABSTRACT

Phencyclidine (PCP) and ketamine are dissociative anesthetics capable of inducing analgesia, psychomimetic behavior, and a catatonic state of unconsciousness. Despite broad similarities, there are notable differences between the clinical actions of ketamine and PCP. Ketamine has a lower incidence of adverse effects and generally produces greater CNS depression than PCP. Both noncompetitively inhibit NMDA receptors, yet there is little evidence that these drugs affect GABA(A) receptors, the primary target of most anesthetics. alpha6beta2/3delta receptors are subtypes of the GABA(A) receptor family and are abundantly expressed in granular neurons within the adult cerebellum. Here, using an oocyte expression system, we show that at anesthetically relevant concentrations, ketamine, but not PCP, modulates alpha6beta2delta and alpha6beta3delta receptors. Additionally, at higher concentrations, ketamine directly activates these GABA(A) receptors. Comparatively, dizocilpine (MK-801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d] cyclohepten-5,10-imine maleate]), a potent noncompetitive antagonist of NMDA receptors that is structurally unrelated to PCP, did not produce any effect on alpha6beta2delta receptors. Of the recombinant GABA(A) receptor subtypes examined (alpha1beta2, alpha1beta2gamma2, alpha1beta2delta, alpha4beta2gamma2, alpha4beta2delta, alpha6beta2gamma2, alpha6beta2delta, and alpha6beta3delta), the actions of ketamine were unique to alpha6beta2delta and alpha6beta3delta receptors. In dissociated granule neurons and cerebellar slice recordings, ketamine potentiated the GABAergic conductance arising from alpha6-containing GABA(A) receptors, whereas PCP showed no effect. Furthermore, ketamine potentiation was absent in cerebellar granule neurons from transgenic functionally null alpha6(-/-) and delta(-/-)mice. These findings suggest that the higher CNS depressant level achieved by ketamine may be the result of its selective actions on alpha6beta2/3delta receptors.


Subject(s)
Cerebellar Cortex/drug effects , Ketamine/pharmacology , Neurons/drug effects , Phencyclidine/pharmacology , Receptors, GABA-A/drug effects , Anesthetics, Dissociative/pharmacology , Animals , Cells, Cultured , Cerebellar Cortex/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Female , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/metabolism , Oocytes , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Xenopus laevis
14.
J Physiol ; 581(Pt 3): 1001-18, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17395622

ABSTRACT

The onset of motor learning in rats coincides with exclusive expression of GABAA receptors containing alpha6 and delta subunits in the granule neurons of the cerebellum. This development temporally correlates with the presence of a spontaneously active chloride current through alpha6-containing GABAA receptors, known as tonic inhibition. Here we report that the coexpression of alpha6, beta2, and delta subunits produced receptor-channels which possessed two distinct and separable states of agonist affinity, one exhibiting micromolar and the other nanomolar affinities for GABA. The high-affinity state was associated with a significant level of spontaneous channel activity. Increasing the level of expression or the ratio of beta2 to alpha6 and delta subunits increased the prevalence of the high-affinity state. Comparative studies of alpha6beta2delta, alpha1beta2delta, alpha6beta2gamma2, alpha1beta2gamma2 and alpha4beta2delta receptors under equivalent levels of expression demonstrated that the significant level of spontaneous channel activity is uniquely attributable to alpha6beta2delta receptors. The pharmacology of spontaneous channel activity arising from alpha6beta2delta receptor expression corresponded to that of tonic inhibition. For example, GABAA receptor antagonists, including furosemide, blocked the spontaneous current. Further, the neuroactive steroid 5alpha-THDOC and classical glycine receptor agonists beta-alanine and taurine directly activated alpha6beta2delta receptors with high potency. Specific mutation within the GABA-dependent activation domain (betaY157F) impaired both low- and high-affinity components of GABA agonist activity in alpha6betaY157Fdelta receptors, but did not attenuate the spontaneous current. In comparison, a mutation located between the second and third transmembrane segments of the delta subunit (deltaR287M) significantly diminished the nanomolar component and the spontaneous activity. The possibility that the high affinity state of the alpha6beta2delta receptor modulates the granule neuron activity as well as potential mechanisms affecting its expression are discussed.


Subject(s)
GABA Agonists/pharmacology , GABA-A Receptor Agonists , Neural Inhibition/drug effects , Neurons/drug effects , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Animals , Bicuculline/pharmacology , Crotonates/pharmacology , Desoxycorticosterone/analogs & derivatives , Desoxycorticosterone/pharmacology , Dose-Response Relationship, Drug , Female , Furosemide/pharmacology , GABA Antagonists/pharmacology , Imidazoles/pharmacology , Microinjections , Mutation , Neurons/metabolism , Oocytes , Picrotoxin/analogs & derivatives , Picrotoxin/pharmacology , Pyridazines/pharmacology , Rats , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Sesterterpenes , Taurine/pharmacology , Xenopus laevis , Zinc/pharmacology , beta-Alanine/pharmacology
15.
Mol Pharmacol ; 66(3): 420-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15322233

ABSTRACT

Fast synaptic transmission in mammalian autonomic ganglia is mediated primarily by nicotinic receptors, and one of the most abundant nicotinic acetylcholine receptor subtypes in these neurons contains the alpha7 subunit (alpha7-nAChRs). Unlike alpha7-nAChRs expressed in other cells, the predominant alpha7-nAChR subtype found in rat intracardiac and superior cervical ganglion neurons exhibits a slow rate of desensitization and is reversibly blocked by alpha-bungarotoxin (alphaBgt). We report here the identification of an alpha7 subunit sequence variant in rat autonomic neurons that incorporates a novel 87-base pair cassette exon in the N terminus of the receptor and preserves the reading frame of the transcript. This alpha7 isoform was detected using reverse transcriptase-polymerase chain reaction techniques in neonatal rat brain and intracardiac and superior cervical ganglion neurons. Immunoblot experiments using a polyclonal antibody directed against the deduced amino acid sequence of the alpha7-2 insert showed a pattern of expression consistent with alpha7-2 subunit mRNA distribution. Moreover, the alpha7-2 subunit could be immunodepleted from protein extracts by solid-phase immunoprecipitation techniques using the anti-alpha7 monoclonal antibody 319. The alpha7-2 subunit was shown to form functional homomeric ion channels that were activated by acetylcholine and blocked by alpha-bungarotoxin when expressed in Xenopus laevis oocytes. This alpha7 isoform exhibited a slow rate of desensitization, and inhibition of these channels by alphaBgt reversed rapidly after washout. Taken together, these data indicate that the alpha7-2 subunit is capable of forming functional alphaBgt-sensitive acetylcholine receptors that resemble the alpha7-nAChRs previously identified in rat autonomic neurons. Furthermore, the distribution of the alpha7-2 isoform is not limited to peripheral neurons.


Subject(s)
Alternative Splicing , Ion Channels/metabolism , Neurons/metabolism , Protein Isoforms/metabolism , Receptors, Nicotinic/metabolism , Acetylcholine/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Molecular Sequence Data , Oocytes/drug effects , Oocytes/metabolism , Protein Isoforms/genetics , Rats , Receptors, Nicotinic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
16.
Arthritis Rheum ; 46(5): 1164-70, 2002 May.
Article in English | MEDLINE | ID: mdl-12115219

ABSTRACT

OBJECTIVE: To compare the efficacy of combination therapy with methotrexate (MTX) and hydroxychloroquine (HCQ), MTX and sulfasalazine (SSZ), and MTX, HCQ, and SSZ in patients with rheumatoid arthritis (RA). METHODS: RA patients (n = 171) who had not previously been treated with combinations of the study medications were randomized to receive 1 of the 3 treatment combinations in this 2-year, double-blind, placebo-controlled protocol. HCQ was given at a dosage of 200 mg twice a day. The dosage of MTX was accelerated from 7.5 mg/week to 17.5 mg/week in all patients who were not in remission. Similarly, the dosage of SSZ was escalated from 500 mg twice a day to 1 gm twice a day in patients who were not in remission. The primary end point of the study was the percentage of patients who had a 20% response to therapy according to the American College of Rheumatology (ACR) criteria at 2 years. RESULTS: Intent-to-treat analysis revealed that patients receiving the triple combination responded best, with 78% achieving an ACR 20% response at 2 years, compared with 60% of those treated with MTX and HCQ (P = 0.05) and 49% of those treated with MTX and SSZ (P = 0.002). Similar trends were seen for the ACR 50% response, with 55%, 40%, and 29% of patients in the 3 treatment groups, respectively, achieving these results at 2 years (P = 0.005 for the triple combination group versus the MTX and SSZ group). All combination treatments were well-tolerated. Fourteen patients (evenly distributed among the 3 groups) withdrew from the protocol because of symptoms that were potentially related to the study medication. CONCLUSION: The triple combination of MTX, SSZ, and HCQ is well-tolerated, and its efficacy is superior to that of the double combination of MTX and SSZ and is marginally superior to that of the double combination of MTX and HCQ.


Subject(s)
Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Hydroxychloroquine/administration & dosage , Methotrexate/administration & dosage , Sulfasalazine/administration & dosage , Adult , Aged , Antirheumatic Agents/adverse effects , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Hydroxychloroquine/adverse effects , Male , Methotrexate/adverse effects , Middle Aged , Sulfasalazine/adverse effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...