Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Auton Neurosci ; 253: 103181, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38696917

ABSTRACT

Respiratory interoception is one of the internal bodily systems that is comprised of different types of somatic and visceral sensations elicited by different patterns of afferent input and respiratory motor drive mediating multiple respiratory modalities. Respiratory interoception is a complex system, having multiple afferents grouped into afferent clusters and projecting into both discriminative and affective centers that are directly related to the behavioral assessment of breathing. The multi-afferent system provides a spectrum of input that result in the ability to interpret the different types of respiratory interceptive sensations. This can result in a response, commonly reported as breathlessness or dyspnea. Dyspnea can be differentiated into specific modalities. These respiratory sensory modalities lead to a general sensation of an Urge-to-Breathe, driven by a need to compensate for the modulation of ventilation that has occurred due to factors that have affected breathing. The multiafferent system for respiratory interoception can also lead to interpretation of the sensory signals resulting in respiratory related sensory experiences, including the Urge-to-Cough and Urge-to-Swallow. These behaviors are modalities that can be driven through the differentiation and integration of multiple afferent input into the respiratory neural comparator. Respiratory sensations require neural somatic and visceral interoceptive elements that include gated attention and detection leading to respiratory modality discrimination with subsequent cognitive decision and behavioral compensation. Studies of brain areas mediating cortical and subcortical respiratory sensory pathways are summarized and used to develop a model of an integrated respiratory neural network mediating respiratory interoception.


Subject(s)
Interoception , Humans , Interoception/physiology , Animals , Respiration , Afferent Pathways/physiology
2.
Article in English | MEDLINE | ID: mdl-38766774

ABSTRACT

Sulfur dioxide (SO2), a common environmental and industrial air pollutant, possesses a potent effect in eliciting cough reflex, but the primary type of airway sensory receptors involved in its tussive action has not been clearly identified. This study was carried out to determine the relative roles of three major types of vagal bronchopulmonary afferents [slowly adapting receptors (SARs), rapidly adapting receptors (RARs), and C-fibers] in regulating the cough response to inhaled SO2. Our results showed that inhalation of SO2 (300 or 600 ppm for 8 min) evoked an abrupt and intense stimulatory effect on bronchopulmonary C-fibers, which continued for the entire duration of inhalation challenge and returned toward the baseline in 1-2 min after resuming room air-breathing in anesthetized and mechanically ventilated mice. In stark contrast, the same SO2 inhalation challenge generated a distinct and consistent inhibitory effect on both SARs and phasic RARs; their phasic discharges synchronized with respiratory cycles during the baseline (breathing room air) began to decline progressively within 1-3 min after the onset of SO2 inhalation, ceased completely after the 8-min inhalation challenge, and then slowly returned toward the baseline after >40 min. In a parallel study in awake mice, inhalation of SO2 at the same concentration and duration as that in the nerve recording experiments evoked cough responses in a pattern and time course similar to that observed in the C-fiber responses. Based upon these results, we concluded that stimulation of vagal bronchopulmonary C-fibers is primarily responsible for triggering the cough response to inhaled SO2.

3.
J Appl Physiol (1985) ; 134(5): 1075-1082, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36958348

ABSTRACT

Slowly adapting receptors (SARs), vagal mechanosensitive receptors located in the lung, play an important role in regulating the breathing pattern and Hering-Breuer inflation reflex (HBIR). Inhalation of high concentration of sulfur dioxide (SO2), a common environmental and occupational air pollutant, has been shown to selectively block the SAR activity in rabbits, but the mechanism underlying this inhibitory effect remained a mystery. We carried out this study to determine if inhalation of SO2 can inhibit the HBIR and change the eupneic breathing pattern, and to investigate further a possible involvement of voltage-gated K+ channels in the inhibitory effect of SO2 on these vagal reflex-mediated responses. Our results showed 1) inhalation of SO2 (600 ppm; 8 min) consistently abolished both the phasic activity of SARs and their response to lung inflation in anesthetized, artificially ventilated mice, 2) inhalation of SO2 generated a distinct inhibitory effect on the HBIR and induced slow deep breathing in anesthetized, spontaneously breathing mice, and these effects were reversible and reproducible in the same animals, 3) This inhibitory effect of SO2 was blocked by pretreatment with 4-aminopyridine (4-AP), a nonselective blocker of voltage-gated K+ channel, and unaffected by pretreatment with its vehicle. In conclusion, this study suggests that this inhibitory effect on the baseline breathing pattern and the HBIR response was primarily mediated through the SO2-induced activation of voltage-gated K+ channels located in the vagal bronchopulmonary SAR neurons.NEW & NOTEWORTHY This study demonstrated that inhaled sulfur dioxide completely and reversibly abolished the activity of vagal bronchopulmonary slowly adapting receptors, significantly inhibited the apneic response to lung inflation, and induced slow deep breathing in anesthetized mice. More importantly, our results further suggested that this inhibitory effect was mediated through an action of sulfur dioxide and its derivatives on the voltage-gated potassium channels expressed in the slowly adapting receptor sensory neurons innervating the lung.


Subject(s)
Potassium Channels, Voltage-Gated , Sulfur Dioxide , Rabbits , Animals , Mice , Sulfur Dioxide/pharmacology , Potassium Channels, Voltage-Gated/pharmacology , Respiration , Lung , Reflex , Vagus Nerve , Apnea , 4-Aminopyridine/pharmacology
4.
Front Physiol ; 13: 813096, 2022.
Article in English | MEDLINE | ID: mdl-35480033

ABSTRACT

Mechanosensitive vagal afferents in the lung, rapidly and slowly adapting receptors (RARs and SARs, respectively), play an important role in eliciting the reflexes that regulate the normal airway function. A profound bronchoconstrictive effect of 5-hydroxytryptamine (5-HT) has been extensively reported in various animal species, but its influence on the SAR and RAR activity is not known. This study investigated the effect of 5-HT on these receptors, and the possible mechanisms involved. Single-fiber activities of these afferents were measured in anesthetized, open-chest, and mechanically ventilated rats. Our results showed that intravenous injection of 5-HT evoked a consistent and pronounced stimulation of phasic RARs. In contrast, 5-HT generated an inconsistent and paradoxical action on SARs: no effect in 29% (5 of 17) of the SARs; stimulation in 35% (6 of 17); and inhibition in the remainder. These responses of both RARs and SARs to 5-HT were reproducible and dose-dependent. After the injection of a high dose of 5-HT (16 µg/kg), the receptor responses slowly reached a peak (after ∼8 s) and returned toward the baseline in ∼20 s, accompanied by a consistent increase in total pulmonary resistance and a decrease in dynamic lung compliance in a temporal pattern very similar to the increased receptor activity. When these changes in lung mechanics induced by 5-HT were prevented by pretreatment with salbutamol, a ß2 adrenergic receptor agonist, the delayed responses of both RARs and SARs to 5-HT were also abolished, except that the immediate stimulatory effect on a subset of RARs, the silent RARs, was not affected. In conclusion, 5-HT generated a delayed stimulatory effect on RARs and a paradoxical effect on SARs, which resulted primarily from the 5-HT-induced changes in mechanical properties of the lung.

5.
Front Med (Lausanne) ; 8: 650637, 2021.
Article in English | MEDLINE | ID: mdl-34395460

ABSTRACT

Extensive evidence indicates that several types of temperature-sensitive ion channels are abundantly expressed in the sensory nerves innervating airway mucosa. Indeed, airway temperature is known to play an important role in regulating respiratory functions. However, the actual airway mucosal temperature and its dynamic changes during the respiratory cycle have not been directly measured. In previous studies, airway tissue temperature was often estimated by indirect measurement of the peak exhaled breath temperature (PEBT). In view of the poor thermal conductivity of air, we believe that the airway tissue temperature cannot be accurately determined by the exhaled air temperature, and this study aimed to test this hypothesis. We applied a miniature rapid-response temperature probe to measure directly the mucosal temperatures of trachea, major, lobar, and segmental bronchi in eight human subjects during a bronchoscopy procedure. Unlike the air temperature in the airway lumen, the mucosal temperature in these airway segments remained relatively stable and did not exhibit the phasic changes synchronous with respiratory cycles. The airway mucosal temperature increased progressively from the extra-thoracic trachea (35.7 ± 0.2°C) toward the segmental bronchus (36.9 ± 0.2°C). Most importantly, the temperatures measured directly at the mucosa of all these airway segments were substantially higher than the PEBT (31.7 ± 0.8°C). The recent findings of a close association between an increased PEBT and airway tissue inflammation have revealed the implication and potential of incorporating the PEBT measurement in the future clinical diagnosis of airway inflammation. Therefore, it is imperative to recognize this distinct difference in temperature between airway mucosa and exhaled air.

6.
Neurosci Lett ; 748: 135719, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33587987

ABSTRACT

Transient Receptor Potential (TRP) channels expressed in specific subsets of airway sensory nerves function as transducers and integrators of a diverse range of sensory inputs including chemical, mechanical and thermal signals. These TRP sensors can detect inhaled irritants as well as endogenously released chemical substances. They play an important role in generating the afferent activity carried by these sensory nerves and regulating the centrally mediated pulmonary defense reflexes. Increasing evidence reported in recent investigations has revealed important involvements of several TRP channels (TRPA1, TRPV1, TRPV4 and TRPM8) in the manifestation of various symptoms and pathogenesis of certain acute and chronic airway diseases. This mini-review focuses primarily on these recent findings of the responses of these TRP sensors to the biological stresses emerging under the pathophysiological conditions of the lung and airways.


Subject(s)
Afferent Pathways/physiology , Lung/physiology , Sensory Receptor Cells/physiology , Transient Receptor Potential Channels/physiology , Animals , Humans , Lung/innervation , Peripheral Nervous System , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology
7.
J Physiol ; 598(5): 1093-1108, 2020 03.
Article in English | MEDLINE | ID: mdl-31891193

ABSTRACT

KEY POINTS: Brief inhalation of SO2 of concentration >500 p.p.m. triggered a pronounced stimulatory effect on vagal bronchopulmonary C-fibres in anaesthetized rats. This stimulatory effect was drastically diminished by a pretreatment with NaHCO3 that raised the baseline arterial pH, suggesting a possible involvement of acidification of airway fluid and/or tissue generated by inhaled SO2 . The stimulation was completely abolished by pretreatment with antagonists of both acid-sensing ion channels and transient receptor potential vanilloid type-1 receptors, indicating that this effect was caused by acid activation of these cation channels expressed in airway sensory nerves. This conclusion was further supported by the results obtained from studies in isolated rat vagal bronchopulmonary sensory neurones and also in the cough response to SO2 inhalation challenge in awake mice. These results provide new insight into the underlying mechanism of harmful irritant effects in the respiratory tract caused by accidental exposure to a high concentration of SO2 . ABSTRACT: Inhalation of sulfur dioxide (SO2 ) triggers coughs and reflex bronchoconstriction, and stimulation of vagal bronchopulmonary C-fibres is primarily responsible. However, the mechanism underlying this stimulatory effect is not yet fully understood. In this study, we tested the hypothesis that the C-fibre stimulation was caused by SO2 -induced local tissue acidosis in the lung and airways. Single-unit activities of bronchopulmonary C-fibres in response to inhalation challenges of SO2 (500-1500 p.p.m., 10 breaths) were measured in anaesthetized rats. Inhalation of SO2 reproducibly induced a pronounced and sustained stimulation (lasting for 15-60 s) of pulmonary C-fibres in a concentration-dependent manner. This stimulatory effect was significantly attenuated by an increase in arterial pH generated by infusion of sodium bicarbonate (NaHCO3 ), and completely abrogated by a combined pretreatment with amiloride (an antagonist of acid-sensing ion channels, ASICs) and AMG8910 (a selective antagonist of the transient receptor potential vanilloid type-1 receptor, TRPV1). Furthermore, in isolated rat vagal pulmonary sensory neurones, perfusion of an aqueous solution of SO2 evoked a transient increase in the intracellular Ca2+ concentration; this response was also markedly diminished by a pretreatment with amiloride and AMG8910. In addition, inhalation of SO2 consistently evoked coughs in awake mice; responses were significantly smaller in TRPV1-/- mice than in wild-type mice, and almost completely abolished after a pretreatment with amiloride in TRPV1-/- mice. These results suggested that the stimulatory effect of inhaled SO2 on bronchopulmonary C-fibres was generated by acidification of fluid and/or tissue in the lung and airways, which activated both ASICs and TRPV1 expressed in these sensory nerves.


Subject(s)
Bronchi , Sulfur Dioxide , Animals , Lung , Mice , Nerve Fibers, Unmyelinated , Rats , Sulfur Dioxide/toxicity , TRPV Cation Channels , Vagus Nerve
8.
Front Physiol ; 10: 642, 2019.
Article in English | MEDLINE | ID: mdl-31191346

ABSTRACT

5-hydroxytryptamine (5-HT) is an inflammatory mediator known to be released in lung. Capsaicin-sensitive lung vagal (CSLV) afferents function as a primary sensor for detecting chemical stimuli and produce consequent reflexes during lung inflammation. To characterize the effect of 5-HT on CSLV afferents, responses of cardiorespiratory reflexes and single-unit C-fiber afferents to right-atrial injections of 5-HT were investigated in anesthetized Sprague-Dawley rats. Bolus injection of 5-HT (8 µg/kg) caused an immediate augmented breath and apnea, accompanied by hypotension and bradycardia. These initial responses were then followed by a brief pressor response and a more sustained depressor response. After a perineural treatment of both cervical vagi with capsaicin to block the conduction of C fibers, 5-HT still triggered the augmented breath, but no longer evoked the apnea, bradycardia and hypotension, indicating an involvement of C-fiber activation. The remaining augmented breath induced by 5-HT after perineural capsaicin treatment was totally eliminated by vagotomy. To further study the effect of 5-HT on CSLV afferents, activities arising from these afferents were determined using the single-fiber recording technique. Right-atrial injection of 5-HT evoked an intense discharge in CSLV afferents in a dose-dependent manner. The highest dose of 5-HT (16 µg/kg) activated 79% (19/24) of CSLV afferents which were also sensitive to capsaicin (0.8 µg/kg). The pretreatment of tropisetron, a selective antagonist of the 5-HT3 receptor, completely blocked CSLV-afferents stimulation induced by 5-HT but did not affect that by capsaicin. Furthermore, a similar afferent response of CSLV afferents was mimicked by phenylbiguanide, a selective agonist of the 5-HT3 receptor. In isolated rat lung vagal C neurons, 5-HT induced intense calcium transients in a dose-dependent manner. The highest concentration (3 µM) of 5-HT activated 67% (18/27) of the CSLV neurons. The 5-HT-induced response was totally abolished by pretreatment of tropisetron. In conclusion, 5-HT exerts an intense stimulatory effect on lung C-fiber terminals mediated through an activation of the 5-HT3 receptor, which may contribute to the airway hypersensitivity under lung inflammation.

9.
Pulm Pharmacol Ther ; 57: 101804, 2019 08.
Article in English | MEDLINE | ID: mdl-31096035

ABSTRACT

Vagal bronchopulmonary C-fiber sensory nerves play an important role in the manifestation of airway hypersensitivity, a common and prominent pathophysiological feature of airway inflammatory diseases. Eosinophil granule-derived cationic proteins are known to be involved in the mucosal damage and development of bronchial hyperresponsiveness during allergic airway inflammation. In view of these background information, we have carried out a series of studies to investigate the effect of cationic proteins on these C-fiber afferents and the mechanism(s) possibly involved; a summary of these studies is presented in this mini-review. Intra-tracheal instillation of either eosinophil granule-derived (e.g., major basic protein, MBP) or synthetic cationic proteins (e.g., poly-l-lysine) induced a sporadic, but intense and lingering discharge of pulmonary C-fibers, and greatly enhanced the chemical and mechanical sensitivities of these afferents in anesthetized rats. The stimulatory and sensitizing effects of these proteins were completely nullified when their cationic charges were neutralized or removed. Furthermore, in isolated rat bronchopulmonary capsaicin-sensitive neurons, eosinophil granule cationic proteins induced a direct and long-lasting (>60 min) but reversible sensitizing effect on their responses to chemical and electrical stimulations. More importantly, our study showed that these cationic proteins exerted an inhibitory effect on the sustained delayed-rectifier voltage-gated K+ current and the A-type, fast-inactivating K+ current; these actions were at least in part responsible for the sensitizing effect in these neurons. In awake mice, intra-tracheal instillation of MBP also induced a slowly developing (peaking in 2-3 days), progressive and sustained (lasting for 3-7 days) elevation of the cough responses to inhaled irritant gases. Taken together, these findings suggest that the enhanced sensitivity of bronchopulmonary C-fibers induced by the eosinophil granule cationic proteins may be a contributing factor in the pathogenesis of bronchial hyperresponsiveness and chronic cough associated with eosinophilic infiltration of the airways.


Subject(s)
Bronchial Hyperreactivity/physiopathology , Cough/physiopathology , Eosinophil Cationic Protein/physiology , Lung/innervation , Vagus Nerve/physiology , Animals , Capsaicin/pharmacology , Cations , Eosinophil Major Basic Protein/pharmacology , Eosinophils/drug effects , Humans , Hypersensitivity/physiopathology , Lung/physiology , Mice , Nerve Fibers, Unmyelinated/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Vagus Nerve Stimulation
10.
Am J Physiol Regul Integr Comp Physiol ; 317(1): R93-R97, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30995073

ABSTRACT

A distinct association between airway eosinophilia and chronic cough is well documented. Eosinophil granule-derived cationic proteins, such as major basic protein (MBP), have been shown to activate and enhance the excitability of bronchopulmonary C-fiber sensory nerves, which may then lead to an increase in cough sensitivity. This study was carried out to determine whether cough responses to inhaled irritant gases were altered by delivery of MBP into the airways. An awake mouse moved freely in a recording chamber that was ventilated with a constant flow of air or irritant gas mixture. Cough responses to separate inhalation challenges of sulfur dioxide (SO2; 300 and 600 ppm) and ammonia (NH3; 0.1 and 0.2%), each for 5-min duration, were measured daily for 3 days before and for up to 8 days after MBP (10-20 µg) instillation into the trachea. During control, inhalations of SO2 and NH3 consistently elicited cough responses in a dose-dependent manner. After MBP treatment, cough responses to both SO2 and NH3 increased significantly and progressively and reached peaks 2-3 days after the treatment before returning to control level in 3-7 days. In sharp contrast, cough responses to these irritant gases were not affected by the treatment with the vehicle of MBP. These results suggest that the MBP-induced lingering elevation of cough responsiveness may be a contributing factor in the pathogenesis of chronic cough associated with eosinophilic infiltration of the airways.


Subject(s)
Ammonia/toxicity , Cough/chemically induced , Eosinophil Major Basic Protein/pharmacology , Sulfur Dioxide/toxicity , Administration, Inhalation , Ammonia/administration & dosage , Animals , Irritants/administration & dosage , Irritants/toxicity , Mice , Respiratory Physiological Phenomena , Sulfur Dioxide/administration & dosage , Wakefulness
11.
JCI Insight ; 4(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30721152

ABSTRACT

Increased airway vagal sensory C-fiber activity contributes to the symptoms of inflammatory airway diseases. The KCNQ/Kv7/M-channel is a well-known determinant of neuronal excitability, yet whether it regulates the activity of vagal bronchopulmonary C-fibers and airway reflex sensitivity remains unknown. Here we addressed this issue using single-cell RT-PCR, patch clamp technique, extracellular recording of single vagal nerve fibers innervating the mouse lungs, and telemetric recording of cough in free-moving mice. Single-cell mRNA analysis and biophysical properties of M-current (IM) suggest that KCNQ3/Kv7.3 is the major M-channel subunit in mouse nodose neurons. The M-channel opener retigabine negatively shifted the voltage-dependent activation of IM, leading to membrane hyperpolarization, increased rheobase, and suppression of both evoked and spontaneous action potential (AP) firing in nodose neurons in an M-channel inhibitor XE991-sensitive manner. Retigabine also markedly suppressed the α,ß-methylene ATP-induced AP firing in nodose C-fiber terminals innervating the mouse lungs, and coughing evoked by irritant gases in awake mice. In conclusion, KCNQ/M-channels play a role in regulating the excitability of vagal airway C-fibers at both the cell soma and nerve terminals. Drugs that open M-channels in airway sensory afferents may relieve the sufferings associated with pulmonary inflammatory diseases such as chronic coughing.


Subject(s)
Cough/metabolism , KCNQ Potassium Channels/metabolism , Vagus Nerve/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anthracenes/pharmacology , Carbamates/pharmacology , KCNQ Potassium Channels/drug effects , KCNQ Potassium Channels/genetics , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/metabolism , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Nerve Tissue Proteins/genetics , Nodose Ganglion , Patch-Clamp Techniques , Phenylenediamines/pharmacology , RNA, Messenger , Transcriptome
12.
Pulm Pharmacol Ther ; 47: 29-37, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28587842

ABSTRACT

Tumor necrosis factor alpha (TNFα) plays a significant role in the pathogenesis of airway inflammatory diseases. Inhalation of aerosolized TNFα induced airway hyperresponsiveness accompanied by airway inflammation in healthy human subjects, but the underlying mechanism is not fully understood. We recently reported a series of studies aimed to investigate if TNFα elevates the sensitivity of vagal bronchopulmonary sensory nerves in a mouse model; these studies are summarized in this mini-review. Our results showed that intratracheal instillation of TNFα induced pronounced airway inflammation 24 h later, as illustrated by infiltration of eosinophils and neutrophils and the release of inflammatory mediators and cytokines in the lung and airways. Accompanying these inflammatory reactions, the sensitivity of vagal pulmonary C-fibers and silent rapidly adapting receptors to capsaicin, a selective agonist of transient receptor potential vanilloid type 1 receptor, was markedly elevated after the TNFα treatment. A distinct increase in the sensitivity to capsaicin induced by TNFα was also observed in isolated pulmonary sensory neurons, suggesting that the sensitizing effect is mediated primarily through a direct action of TNFα on these neurons. Furthermore, the same TNFα treatment also induced a lingering (>7days) cough hyperresponsiveness to inhalation challenge of NH3 in awake mice. Both the airway inflammation and the sensitizing effect on pulmonary sensory neurons caused by the TNFα treatment were abolished in the TNF-receptor double homozygous mutant mice, indicating the involvement of TNF-receptor activation. These findings suggest that the TNFα-induced hypersensitivity of vagal bronchopulmonary afferents may be responsible for, at least in part, the airway hyperresponsiveness caused by inhaled TNFα in healthy individuals.


Subject(s)
Lung/physiopathology , Respiratory Hypersensitivity/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cough/physiopathology , Disease Models, Animal , Humans , Inflammation/physiopathology , Mice , Receptors, Tumor Necrosis Factor/metabolism , Sensory Receptor Cells/metabolism , Vagus Nerve/metabolism
13.
Front Physiol ; 8: 411, 2017.
Article in English | MEDLINE | ID: mdl-28659824

ABSTRACT

Tumor necrosis factor alpha (TNFα), a pro-inflammatory cytokine, plays a significant role in the pathogenesis of allergic asthma. Inhalation of TNFα also induces airway hyperresponsiveness in healthy human subjects, and the underlying mechanism is not fully understood. A recent study reported that TNFα caused airway inflammation and a sustained elevation of pulmonary chemoreflex responses in mice, suggesting a possible involvement of heightened sensitivity of vagal pulmonary C-fibers. To investigate this possibility, the present study aimed to investigate the effect of a pretreatment with TNFα on the sensitivity of vagal pulmonary afferents in anesthetized mice. After TNFα (10 µg/ml, 0.03 ml) and vehicle (Veh; phosphate buffered saline (PBS), 0.03 ml) were administered by intra-tracheal instillation in each mouse of treated (TNF) and control (Veh) groups, respectively, the peak activity of pulmonary C-fibers in response to an intravenous bolus injection of a low dose of capsaicin (Cap; 0.5 µg/kg) was significantly elevated in TNF group (6.5 ± 1.3 impulses/s, n = 12) 24-48 h later, compared to that in Veh group (2.2 ± 0.5 impulses/s, n = 11; P < 0.05). Interestingly, the same low dose of Cap injection also evoked a distinct burst of discharge (2.4 ± 0.7 impulses/s) in 75% of the silent rapidly adapting receptors (RARs), a subtype of RARs exhibiting no phasic activity, in TNF group, but did not stimulate any of the silent RARs in Veh group. To further determine if this sensitizing effect involves a direct action of TNFα on these sensory nerves, the change in intracellular Ca2+ concentration in response to Cap challenge was measured in isolated mouse vagal pulmonary sensory neurons. The Cap-evoked Ca2+ influx was markedly enhanced in the neurons incubated with TNFα (50 ng/ml) for ~24 h, and this sensitizing effect was attenuated in the neurons isolated from the TNF-receptor double homozygous mutant mice. In conclusion, the TNFα pretreatment enhanced the Cap sensitivity in both pulmonary C-fibers and silent RARs, and the action was mediated through TNF receptors. These sensitizing effects of TNFα may contribute, at least in part, to the pathogenesis of airway hyperresponsiveness induced by this cytokine.

14.
Am J Physiol Lung Cell Mol Physiol ; 313(2): L293-L304, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28522561

ABSTRACT

We studied acute effects of tumor necrosis factor-α (TNFα) on the sensitivity of isolated rat vagal pulmonary sensory neurons. Our results showed the following. First, a brief pretreatment with a low dose of TNFα (1.44 nM, 9 min) enhanced the sensitivity of transient receptor potential vanilloid type 1 (TRPV1) receptors in these neurons in two distinct phases: the inward current evoked by capsaicin was amplified (Δ = 247%) immediately following the TNFα pretreatment, which gradually declined toward control and then increased again reaching another peak (Δ = 384%) after 60-90 min. Second, the immediate phase of this potentiating effect of TNFα was completely abolished by a pretreatment with a selective cyclooxygenase-2 (COX-2) inhibitor, NS-398, whereas the delayed potentiation was only partially attenuated. Third, in sharp contrast, TNFα did not generate any potentiating effect on the responses to non-TRPV1 chemical activators of these neurons. Fourth, the selectivity of the TNFα action on TRPV1 was further illustrated by the responses to acid (pH 6.0); TNFα did not affect the rapid transient current mediated by acid-sensing ion channels but significantly augmented the slow sustained current mediated by TRPV1 in the same neurons. Fifth, in anesthetized rats, a similar pattern of acute sensitizing effects of TNFα on pulmonary C-fiber afferents and the involvement of COX-2 were also clearly shown. In conclusion, a brief pretreatment with TNFα induced both immediate and delayed potentiating effects on the TRPV1 sensitivity in pulmonary sensory neurons, and the production of COX-2 arachidonic acid metabolites plays a major role in the immediate sensitizing effect of TNFα.


Subject(s)
Lung/metabolism , Sensory Receptor Cells/metabolism , TRPV Cation Channels/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acid Sensing Ion Channels/metabolism , Animals , Capsaicin/pharmacology , Cyclooxygenase 2/metabolism , Lung/drug effects , Male , Nerve Fibers, Unmyelinated/metabolism , Nitrobenzenes/pharmacology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sulfonamides/pharmacology , Vagus Nerve/drug effects , Vagus Nerve/metabolism
15.
Am J Physiol Regul Integr Comp Physiol ; 312(5): R718-R726, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28228416

ABSTRACT

This study was designed to determine the effect of active sensitization with ovalbumin (Ova) on cough responses to inhaled irritant gases in mice. Conscious mice moved freely in a recording chamber, while the pressure change in the chamber and audio and video signals of the mouse movements were recorded simultaneously to measure the frequencies of cough reflex (CR) and expiration reflex (ER). To further verify the accuracy of cough analysis, the intrapleural pressure was also recorded by a telemetry sensor surgically implanted in the intrapleural space in a subgroup of mice. During the irritant gas inhalation challenge, sulfur dioxide (SO2; 200 and 400 ppm) or ammonia (NH3; 0.1% and 0.2%) was drawn into the chamber at a constant flow rate for 8 min. Ova sensitization and sham sensitization with vehicle (Veh) were performed over a 25-day period in separate groups of mice. Our results showed that 1) both SO2 and NH3 inhalation challenges increased CR and ER frequencies in a concentration-dependent manner before Ova sensitization; 2) the baseline CR frequency was significantly elevated after Ova sensitization, accompanied by pronounced airway inflammation; and 3) Ova sensitization also markedly augmented the responses of CR and ER to both SO2 and NH3 inhalation challenges; in sharp contrast, the cough responses did not change after sham sensitization in the Veh group. In conclusion, Ova sensitization caused distinct and lingering increases in baseline cough frequency, and also intensified both CR and ER responses to inhaled irritant gases, which probably resulted from an allergic inflammation-induced hypersensitivity of airway sensory nerves.


Subject(s)
Cough/physiopathology , Exhalation/drug effects , Lung Injury/chemically induced , Lung Injury/physiopathology , Pneumonia/physiopathology , Reflex/drug effects , Vehicle Emissions/poisoning , Administration, Inhalation , Ammonia/administration & dosage , Ammonia/poisoning , Animals , Inhalation Exposure/adverse effects , Irritants/administration & dosage , Male , Mice , Mice, Inbred C57BL , Ovalbumin , Pneumonia/chemically induced , Pneumonia/complications , Reflex, Abnormal , Sulfur Dioxide/administration & dosage , Sulfur Dioxide/poisoning
16.
Toxicol Sci ; 153(1): 103-11, 2016 09.
Article in English | MEDLINE | ID: mdl-27329243

ABSTRACT

Maternal cigarette smoke is the major risk of sudden infant death syndrome (SIDS). A depressed ventilatory response to hypoxia (HVR) and hypercapnia (HCVR) is thought to be responsible for the pathogenesis of SIDS and the carotid body is critically involved in these responses. We have recently reported that prenatal nicotinic exposure (PNE) over the full gestation induces depressed HVR in rat pups. Here, we asked whether PNE (1) depressed not only HVR but also HCVR that were dependent on the carotid body, (2) affected some important receptors and neurochemicals expressed in the carotid body, such as tyrosine hydroxylase (TH), neurokinin-1 receptor (NK1R), and α7 nicotinic acetylcholine receptor (α7nAChR), and (3) blunted the ventilatory responses to activation of these receptors. To this end, HVR and HCVR in Ctrl and PNE pups were measured with plethysmography before and after carotid body ablation (Series I), mRNA expression and/or immunoreactivity (IR) of TH, NK1R, and α7nAChR in the carotid body were examined by RT-PCR and immunohistochemistry (Series II), and the ventilatory responses were tested before and after intracarotid injection of substance P (NK1R agonist) and AR-R17779 (α7nAChR agonist) (Series III). Our results showed that PNE (1) significantly depressed both HVR and HCVR and these depressions were abolished by carotid body ablation, (2) reduced the relative population of glomus cells, mRNA NK1R, and α7nAChR and IR of NK1R and TH in the carotid body, and (3) decreased ventilatory responses to intracarotid injection of substance P or AR-R17779. These results suggest that PNE acting via the carotid body could strikingly blunt HVR and HCVR, likely through downregulating TH and NK1R.


Subject(s)
Carotid Body/drug effects , Down-Regulation/drug effects , Nicotine/administration & dosage , Prenatal Exposure Delayed Effects , Receptors, Neurokinin-1/metabolism , Respiratory System/drug effects , Tyrosine 3-Monooxygenase/metabolism , Animals , Carotid Body/metabolism , Female , Male , Nicotine/pharmacology , Plethysmography , Pregnancy , Rats , Rats, Sprague-Dawley , alpha7 Nicotinic Acetylcholine Receptor/metabolism
17.
Toxicol Appl Pharmacol ; 290: 107-15, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26524655

ABSTRACT

Prenatal nicotinic exposure (PNE) prolongs bronchopulmonary C-fiber (PCF)-mediated apneic response to intra-atrial bolus injection of capsaicin in rat pups. The relevant mechanisms remain unclear. Pulmonary substance P and adenosine and their receptors (neurokinin-A receptor, NK1R and ADA1 receptor, ADA1R) and transient receptor potential cation channel subfamily V member 1 (TRPV1) expressed on PCFs are critical for PCF sensitization and/or activation. Here, we compared substance P and adenosine in BALF and NK1R, ADA1R, and TRPV1 expression in the nodose/jugular (N/J) ganglia (vagal pulmonary C-neurons retrogradely labeled) between Ctrl and PNE pups. We found that PNE failed to change BALF substance P and adenosine content, but significantly upregulated both mRNA and protein TRPV1 and NK1R in the N/J ganglia and only NK1R mRNA in pulmonary C-neurons. To define the role of NK1R in the PNE-induced PCF sensitization, the apneic response to capsaicin (i.v.) without or with pretreatment of SR140333 (a peripheral and selective NK1R antagonist) was compared and the prolonged apnea by PNE significantly shortened by SR140333. To clarify if the PNE-evoked responses depended on action of nicotinic acetylcholine receptors (nAChRs), particularly α7nAChR, mecamylamine or methyllycaconitine (a general nAChR or a selective α7nAChR antagonist) was administrated via another mini-pump over the PNE period. Mecamylamine or methyllycaconitine eliminated the PNE-evoked mRNA and protein responses. Our data suggest that PNE is able to elevate PCF NK1R expression via activation of nAChRs, especially α7nAChR, which likely contributes to sensitize PCFs and prolong the PCF-mediated apneic response to capsaicin.


Subject(s)
Lung/drug effects , Nerve Fibers, Unmyelinated/drug effects , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-2/metabolism , Up-Regulation , Adenosine/pharmacology , Animals , Animals, Newborn , Apnea/drug therapy , Bronchoalveolar Lavage Fluid , Capsaicin/pharmacology , Female , Ganglia/drug effects , Ganglia/metabolism , Male , Nerve Fibers, Unmyelinated/metabolism , Nicotine/blood , Nicotine/toxicity , Pregnancy , Prenatal Exposure Delayed Effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-2/genetics , Substance P/pharmacology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Vagus Nerve/drug effects , Vagus Nerve/metabolism
18.
Am J Physiol Regul Integr Comp Physiol ; 309(10): R1285-91, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26333786

ABSTRACT

Our recent study has shown that hyperventilation of humidified warm air (HWA) triggered cough and reflex bronchoconstriction in patients with mild asthma. We suggested that a sensitizing effect on bronchopulmonary C-fibers by increasing airway temperature was involved, but direct evidence was lacking. This study was carried out to test the hypothesis that HWA enhances the pulmonary C-fiber sensitivity in Brown-Norway rats sensitized with ovalbumin (Ova). In anesthetized rats, isocapnic hyperventilation of HWA for 3 min rapidly elevated airway temperature to a steady state of 41.7°C. Immediately after the HWA challenge, the baseline fiber activity (FA) of pulmonary C-fibers was markedly elevated in sensitized rats, but not in control rats. Furthermore, the response of pulmonary C-fibers to right atrial injection of capsaicin in sensitized rats was significantly higher than control rats before the HWA challenge, and the response to capsaicin was further amplified after HWA in sensitized rats (ΔFA = 4.51 ± 1.02 imp/s before, and 9.26 ± 1.74 imp/s after the HWA challenge). A similar pattern of the HWA-induced potentiation of the FA response to phenylbiguanide, another chemical stimulant of C-fibers, was also found in sensitized rats. These results clearly demonstrated that increasing airway temperature significantly elevated both the baseline activity and responses to chemical stimuli of pulmonary C-fibers in Ova-sensitized rats. In conclusion, this study supports the hypothesis that the increased excitability of these afferents may have contributed to the cough and reflex bronchoconstriction evoked by hyperventilation of HWA in patients with asthma.


Subject(s)
Hot Temperature , Lung/innervation , Ovalbumin/immunology , Respiratory Hypersensitivity/chemically induced , Vagus Nerve/drug effects , Animals , Humidity , Male , Random Allocation , Rats
19.
Pulm Pharmacol Ther ; 35: 87-93, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26283426

ABSTRACT

Transient receptor potential ankyrin type 1 (TRPA1) and vanilloid type 1 (TRPV1) receptors are co-expressed in vagal pulmonary C-fiber sensory nerves. Because both these ligand-gated non-selective cation channels are sensitive to a number of endogenous inflammatory mediators, it is highly probable that they can be activated simultaneously during airway inflammation. Studies were carried out to investigate whether there is an interaction between these two polymodal transducers upon simultaneous activation, and how it modulates the activity of vagal pulmonary C-fiber sensory nerves. Our studies showed a distinct potentiating effect induced abruptly by simultaneous activations of TRPA1 and TRPV1 by their respective selective agonists, allyl isothiocyanate (AITC) and capsaicin (Cap), at near-threshold concentrations. This synergistic effect was demonstrated in the studies of single-unit recording of vagal bronchopulmonary C-fiber afferents and the reflex responses elicited by activation of these afferents in intact animals, as well as in the isolated nodose and jugular bronchopulmonary sensory neurons. This potentiating effect was absent when either AITC or Cap was replaced by non-TRPA1 and non-TRPV1 chemical activators of these neurons, demonstrating the selectivity of the interaction between these two TRP channels. Furthermore, the synergism was dependent upon the extracellular Ca(2+), and the rapid onset of the action further suggests that the interaction probably occurred locally at the sites of these channels. These findings suggest that the TRPA1-TRPV1 interaction may play an important role in regulating the function and excitability of pulmonary sensory neurons during airway inflammation, but the mechanism underlying this positive interaction is not yet fully understood.


Subject(s)
Calcium Channels/physiology , Lung/innervation , Nerve Tissue Proteins/physiology , Sensory Receptor Cells/physiology , TRPV Cation Channels/genetics , Transient Receptor Potential Channels/physiology , Animals , Calcium Channels/genetics , Humans , Nerve Fibers, Unmyelinated , Nerve Tissue Proteins/genetics , Pneumonia/physiopathology , TRPA1 Cation Channel , TRPV Cation Channels/physiology , Transient Receptor Potential Channels/genetics
20.
J Appl Physiol (1985) ; 118(12): 1533-43, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25858491

ABSTRACT

Both transient receptor potential ankyrin 1 (TRPA1) and vanilloid 1 (TRPV1) receptors are abundantly expressed in bronchopulmonary C-fiber sensory nerves and can be activated by a number of endogenous inflammatory mediators. A recent study has reported a synergistic effect of simultaneous TRPA1 and TRPV1 activations in vagal pulmonary C-fiber afferents in anesthetized rats, but its underlying mechanism was not known. This study aimed to characterize a possible interaction between these two TRP channels and to investigate the potential role of Ca(2+) as a mediator of this interaction in isolated rat vagal pulmonary sensory neurons. Using the perforated patch-clamp recording technique, our study demonstrated a distinct positive interaction occurring abruptly between TRPA1 and TRPV1 when they were activated simultaneously by their respective agonists, capsaicin (Cap) and allyl isothiocyanate (AITC), at near-threshold concentrations in these neurons. AITC at this low concentration evoked only minimal or undetectable responses, but it markedly amplified the Cap-evoked current in the same neurons. This potentiating effect was eliminated when either AITC or Cap was replaced by non-TRPA1 and non-TRPV1 chemical activators of these neurons, demonstrating the selectivity of the interaction between these two TRP channels. Furthermore, when Ca(2+) was removed from the extracellular solution, the synergistic effect of Cap and AITC on pulmonary sensory neurons was completely abrogated, clearly indicating a critical role of Ca(2+) in mediating the action. These results suggest that this TRPA1-TRPV1 interaction may play a part in regulating the sensitivity of pulmonary sensory neurons during airway inflammatory reaction.


Subject(s)
Bronchi/innervation , Calcium/physiology , Lung/innervation , Sensory Receptor Cells/physiology , TRPC Cation Channels/physiology , TRPV Cation Channels/physiology , Animals , Calcium Channel Agonists/pharmacology , Capsaicin/pharmacology , Drug Synergism , Ions , Male , Nerve Fibers, Unmyelinated/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , TRPA1 Cation Channel , TRPC Cation Channels/drug effects , TRPC Cation Channels/metabolism , TRPV Cation Channels/drug effects , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...