Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Sci Rep ; 11(1): 20823, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675332

ABSTRACT

Peripheral chemoreceptors' (PCh) hyperactivity increases sympathetic tone. An augmented acute ventilatory response to hypoxia, being a marker of PCh oversensitivity, was also identified as a marker of poor prognosis in HF. However, not much is known about the tonic (chronic) influence of PCh on cardio-respiratory parameters. In our study 30 HF patients and 30 healthy individuals were exposed to 100% oxygen for 1 min during which minute ventilation and hemodynamic parameters were non-invasively recorded. Systemic vascular resistance (SVR) and mean arterial pressure (MAP) responses to acute hyperoxia differed substantially between HF and control. In HF hyperoxia caused a significant drop in SVR in early stages with subsequent normalization, while increase in SVR was observed in controls. MAP increased in controls, but remained unchanged in HF. Bilateral carotid bodies excision performed in two HF subjects changed the response to hyperoxia towards the course seen in healthy individuals. These differences may be explained by the domination of early vascular reaction to hyperoxia in HF by vasodilation due to the inhibition of augmented tonic activity of PCh. Otherwise, in healthy subjects the vasoconstrictive action of oxygen remains unopposed. The magnitude of SVR change during acute hyperoxia may be used as a novel method for tonic PCh activity assessment.


Subject(s)
Chemoreceptor Cells/pathology , Heart Failure/complications , Hyperoxia/complications , Vascular Resistance , Aged , Female , Heart Failure/pathology , Heart Failure/physiopathology , Hemodynamics , Humans , Hyperoxia/pathology , Hyperoxia/physiopathology , Male , Middle Aged
2.
Viruses ; 13(8)2021 08 20.
Article in English | MEDLINE | ID: mdl-34452517

ABSTRACT

Patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019, suffer from respiratory and non-respiratory symptoms. Among these symptoms, the loss of smell has attracted considerable attention. The objectives of this study were to determine which cells are infected, what happens in the olfactory system after viral infection, and how these pathologic changes contribute to olfactory loss. For this purpose, Syrian golden hamsters were used. First, we verified the olfactory structures in the nasal cavity of Syrian golden hamsters, namely the main olfactory epithelium, the vomeronasal organ, and their cellular components. Second, we found angiotensin-converting enzyme 2 expression, a receptor protein of SARS-CoV-2, in both structures and infections of supporting, microvillar, and solitary chemosensory cells. Third, we observed pathological changes in the infected epithelium, including reduced thickness of the mucus layer, detached epithelia, indistinct layers of epithelia, infiltration of inflammatory cells, and apoptotic cells in the overall layers. We concluded that a structurally and functionally altered microenvironment influences olfactory function. We observed the regeneration of the damaged epithelium, and found multilayers of basal cells, indicating that they were activated and proliferating to reconstitute the injured epithelium.


Subject(s)
COVID-19/virology , Chemoreceptor Cells/virology , Olfactory Mucosa/virology , SARS-CoV-2 , Vomeronasal Organ/virology , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/pathology , Chemoreceptor Cells/pathology , Male , Mesocricetus , Nasal Cavity/pathology , Nasal Cavity/virology , Olfactory Mucosa/metabolism , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/pathology , Olfactory Receptor Neurons/virology , Receptors, Coronavirus/metabolism , Regeneration , SARS-CoV-2/isolation & purification , Vomeronasal Organ/metabolism , Vomeronasal Organ/pathology
3.
PLoS One ; 15(10): e0237643, 2020.
Article in English | MEDLINE | ID: mdl-33064729

ABSTRACT

We previously reported that maternal cigarette smoke (CS) exposure resulted in impairment of central chemoreception and induced mitochondrial dysfunction in offspring parafacial respiratory group (pFRG), the kernel for mammalian central chemoreception. We also found that hydrogen sulfide (H2S) could attenuate maternal CS exposure-induced impairment of central chemoreception in the rat offspring in vivo. Mitochondrial ATP sensitive potassium (mitoKATP) channel has been reported to play a significant role in mitochondrial functions and protect against apoptosis in neurons. Thus, we hypothesize here that mitoKATP channel plays a role in the protective effects of H2S on neonatal central chemoreception in maternal CS-exposed rats. Our findings revealed that pretreatment with NaHS (donor of H2S, 22.4mM) reversed the central chemosensitivity decreased by maternal CS exposure, and also inhibited cell apoptosis in offspring pFRG, however, 5-HD (blocker of mitoKATP channels, 19mM) attenuated the protective effects of NaHS. In addition, NaHS declined pro-apoptotic proteins related to mitochondrial pathway apoptosis in CS rat offspring pFRG, such as Bax, Cytochrome C, caspase9 and caspase3. NaHS or 5-HD alone had no significant effect on above indexes. These results suggest that mitoKATP channels play an important role in the protective effect of H2S against impairment of central chemoreception via anti-apoptosis in pFRG of rat offspring exposed to maternal CS.


Subject(s)
Chemoreceptor Cells/drug effects , Cigarette Smoking/adverse effects , Hydrogen Sulfide/metabolism , Maternal Exposure/adverse effects , Potassium Channels/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Chemoreceptor Cells/pathology , Chemoreceptor Cells/physiology , Female , Medulla Oblongata/drug effects , Medulla Oblongata/pathology , Medulla Oblongata/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Protective Agents/metabolism , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Respiratory Mechanics/drug effects , Respiratory Mechanics/physiology , Sulfides/metabolism , Sulfides/pharmacology
4.
Gastroenterology ; 159(6): 2101-2115.e5, 2020 12.
Article in English | MEDLINE | ID: mdl-32828819

ABSTRACT

BACKGROUND & AIMS: Countries endemic for parasitic infestations have a lower incidence of Crohn's disease (CD) than nonendemic countries, and there have been anecdotal reports of the beneficial effects of helminths in CD patients. Tuft cells in the small intestine sense and direct the immune response against eukaryotic parasites. We investigated the activities of tuft cells in patients with CD and mouse models of intestinal inflammation. METHODS: We used microscopy to quantify tuft cells in intestinal specimens from patients with ileal CD (n = 19), healthy individuals (n = 14), and TNFΔARE/+ mice, which develop Crohn's-like ileitis. We performed single-cell RNA sequencing, mass spectrometry, and microbiome profiling of intestinal tissues from wild-type and Atoh1-knockout mice, which have expansion of tuft cells, to study interactions between microbes and tuft cell populations. We assessed microbe dependence of tuft cell populations using microbiome depletion, organoids, and microbe transplant experiments. We used multiplex imaging and cytokine assays to assess alterations in inflammatory response following expansion of tuft cells with succinate administration in TNFΔARE/+ and anti-CD3E CD mouse models. RESULTS: Inflamed ileal tissues from patients and mice had reduced numbers of tuft cells, compared with healthy individuals or wild-type mice. Expansion of tuft cells was associated with increased expression of genes that regulate the tricarboxylic acid cycle, which resulted from microbe production of the metabolite succinate. Experiments in which we manipulated the intestinal microbiota of mice revealed the existence of an ATOH1-independent population of tuft cells that was sensitive to metabolites produced by microbes. Administration of succinate to mice expanded tuft cells and reduced intestinal inflammation in TNFΔARE/+ mice and anti-CD3E-treated mice, increased GATA3+ cells and type 2 cytokines (IL22, IL25, IL13), and decreased RORGT+ cells and type 17 cytokines (IL23) in a tuft cell-dependent manner. CONCLUSIONS: We found that tuft cell expansion reduced chronic intestinal inflammation in mice. Strategies to expand tuft cells might be developed for treatment of CD.


Subject(s)
Chemoreceptor Cells/immunology , Crohn Disease/immunology , Gastrointestinal Microbiome/immunology , Ileitis/immunology , Intestinal Mucosa/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Chemoreceptor Cells/pathology , Crohn Disease/microbiology , Crohn Disease/pathology , DNA, Bacterial/genetics , Disease Models, Animal , Feces/microbiology , Female , Humans , Ileitis/microbiology , Ileitis/pathology , Ileum/cytology , Ileum/immunology , Ileum/microbiology , Ileum/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Protective Factors , RNA, Ribosomal, 16S/genetics , RNA-Seq , Single-Cell Analysis , Succinic Acid/immunology , Succinic Acid/metabolism
5.
J Insect Sci ; 20(4)2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32677682

ABSTRACT

Cultures of Manduca sexta Johanssen in our laboratory were found to have larvae with missing or deformed mouthparts or antennae. Hypothesizing that these developmental deformities were caused by crowded rearing conditions, we reared larvae in four different population densities and recorded the incidence (% of larvae affected) and types of chemoreceptor deformities. Results showed that the incidence of these deformities was directly proportional to larval population density. Deformities of the maxilla and palp were the most frequent, followed by those of the antenna, epipharynx and maxillary styloconica. Life history traits of larval mass, food consumption, and rate of development were inversely related to larval density for both normal and deformed larvae. We discuss possible causes and mechanisms of these deformities and of changes to life history traits.


Subject(s)
Chemoreceptor Cells/pathology , Larva/growth & development , Manduca/growth & development , Animals , Population Density
6.
J Neurovirol ; 26(3): 371-381, 2020 06.
Article in English | MEDLINE | ID: mdl-32144727

ABSTRACT

Zika virus (ZIKV) is an emerging virus belonging to the genus Flavivirus. ZIKV infection is a significant health concern, with increasing numbers of reports of microcephaly cases in fetuses and Guillain-Barré syndrome (GBS) in adults. Interestingly, chemosensory disturbances are also reported as one of the manifestations of GBS. ZIKV infects several human tissues and cell types in vitro and in vivo. However, there is no study demonstrating ZIKV infection and replication in chemosensory cells, including olfactory and taste cells. Taste papilla and olfactory cells are chemosensory receptor cells with unique histological, molecular, and physiological characteristics. Here we examined ZIKV infection (PRVABC59) in cultured human olfactory epithelial cells (hOECs) and fungiform taste papilla (HBO) cells in vitro, as well as in vivo mouse taste and olfactory epithelial and olfactory bulb tissues. Interestingly, while HBO cells showed resistance to ZIKV replication, hOECs were highly susceptible for ZIKV infection and replication. Further, we demonstrated the presence of ZIKV particles and expression of viral proteins in olfactory epithelium, as well as in olfactory bulb, but not in taste papillae, of immunocompromised mice (ifnar/-) infected with the PRVABC59 strain of ZIKV. These observations suggest that chemosensory cells in the olfactory neuroepithelium and olfactory bulb may be important tissues for ZIKV replication and dissemination.


Subject(s)
Chemoreceptor Cells/virology , Receptor, Interferon alpha-beta/immunology , Virus Replication/physiology , Zika Virus Infection/virology , Zika Virus/pathogenicity , Animals , Cell Line , Chemoreceptor Cells/immunology , Chemoreceptor Cells/pathology , Female , Gene Expression , Host-Pathogen Interactions/immunology , Humans , Male , Mice , Mice, Knockout , Organ Specificity , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Smell/physiology , Taste/physiology , Zika Virus/growth & development , Zika Virus/immunology , Zika Virus Infection/immunology , Zika Virus Infection/pathology
7.
J Physiol ; 596(13): 2521-2545, 2018 07.
Article in English | MEDLINE | ID: mdl-29667182

ABSTRACT

KEY POINTS: The retrotrapezoid nucleus (RTN) drives breathing proportionally to brain PCO2 but its role during various states of vigilance needs clarification. Under normoxia, RTN lesions increased the arterial PCO2 set-point, lowered the PO2 set-point and reduced alveolar ventilation relative to CO2 production. Tidal volume was reduced and breathing frequency increased to a comparable degree during wake, slow-wave sleep and REM sleep. RTN lesions did not produce apnoeas or disordered breathing during sleep. RTN lesions in rats virtually eliminated the central respiratory chemoreflex (CRC) while preserving the cardiorespiratory responses to hypoxia; the relationship between CRC and number of surviving RTN Nmb neurons was an inverse exponential. The CRC does not function without the RTN. In the quasi-complete absence of the RTN and CRC, alveolar ventilation is reduced despite an increased drive to breathe from the carotid bodies. ABSTRACT: The retrotrapezoid nucleus (RTN) is one of several CNS nuclei that contribute, in various capacities (e.g. CO2 detection, neuronal modulation) to the central respiratory chemoreflex (CRC). Here we test how important the RTN is to PCO2 homeostasis and breathing during sleep or wake. RTN Nmb-positive neurons were killed with targeted microinjections of substance P-saporin conjugate in adult rats. Under normoxia, rats with large RTN lesions (92 ± 4% cell loss) had normal blood pressure and arterial pH but were hypoxic (-8 mmHg PaO2 ) and hypercapnic (+10 mmHg ). In resting conditions, minute volume (VE ) was normal but breathing frequency (fR ) was elevated and tidal volume (VT ) reduced. Resting O2 consumption and CO2 production were normal. The hypercapnic ventilatory reflex in 65% FiO2 had an inverse exponential relationship with the number of surviving RTN neurons and was decreased by up to 92%. The hypoxic ventilatory reflex (HVR; FiO2 21-10%) persisted after RTN lesions, hypoxia-induced sighing was normal and hypoxia-induced hypotension was reduced. In rats with RTN lesions, breathing was lowest during slow-wave sleep, especially under hyperoxia, but apnoeas and sleep-disordered breathing were not observed. In conclusion, near complete RTN destruction in rats virtually eliminates the CRC but the HVR persists and sighing and the state dependence of breathing are unchanged. Under normoxia, RTN lesions cause no change in VE but alveolar ventilation is reduced by at least 21%, probably because of increased physiological dead volume. RTN lesions do not cause sleep apnoea during slow-wave sleep, even under hyperoxia.


Subject(s)
Blood Gas Analysis , Carbon Dioxide/analysis , Chemoreceptor Cells/pathology , Homeostasis , Medulla Oblongata/physiopathology , Pulmonary Ventilation , Respiration , Animals , Hypoxia , Male , Neurokinin B/analogs & derivatives , Neurokinin B/metabolism , Rats , Rats, Sprague-Dawley , Sleep
8.
Neuroscience ; 369: 350-362, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29191463

ABSTRACT

Central chemoreceptors are primarily sensitive to changes in CO2/H+, and such changes lead to intense breathing activity. Medullary raphe and retrotrapezoid nucleus (RTN) neurons are candidates for central chemoreceptors because they are unusually pH sensitive. The pathophysiology of Parkinson's disease (PD) is related to the reduction of neurons in the substantia nigra pars compacta (SNpc) that express dopamine, although other neurons can also be degenerated in this pathology. In rodent models of PD, we showed an impairment of the hypercapnia ventilatory response due to a reduction in the number of RTN chemosensitive neurons. Here, we aimed to investigate if serotonine-expressing neurons in the Raphe pallidus/parapyramidal region (RPa/PPy) are also involved in the modulation of breathing during central chemoreception activation in a PD animal model. PD was induced in male Wistar rats with bilateral injection of 6-OHDA (6-hydroxydopamine; 24 µg/µl) into the striatum, which leads to a reduction in the catecholaminergic neurons of the SNpc by 89%. In PD animals, we noticed a reduction in the number of RPa neurons that project to the RTN, without a change in the number of hypercapnia-activated (7% CO2) raphe neurons. The PD animals that received injection of the toxin saporin anti-SERT into the RPA/PPy region did not show a further reduction of respiratory frequency (fR) or ventilation (VE) at rest or during hypercapnia challenge. These experiments demonstrate that serotonergic neurons of RPa/PPy are not involved in the breathing responses induced by central chemoreceptor activation in a PD animal model.


Subject(s)
Chemoreceptor Cells/physiology , Nucleus Raphe Pallidus/physiopathology , Parkinsonian Disorders/physiopathology , Respiration , Animals , Carbon Dioxide/metabolism , Chemoreceptor Cells/pathology , Hypercapnia/pathology , Hypercapnia/physiopathology , Male , Neural Pathways/pathology , Neural Pathways/physiopathology , Nucleus Raphe Pallidus/pathology , Oxidopamine , Parkinsonian Disorders/pathology , Rats, Wistar , Serotonergic Neurons/pathology , Serotonergic Neurons/physiology
9.
J Nutr ; 145(5): 907-14, 2015 May.
Article in English | MEDLINE | ID: mdl-25934662

ABSTRACT

BACKGROUND: Maternal protein restriction in rats increases the risk of adult offspring arterial hypertension through unknown mechanisms. OBJECTIVES: The aims of the study were to evaluate the effects of a low-protein (LP) diet during pregnancy and lactation on baseline sympathetic and respiratory activities and peripheral chemoreflex sensitivity in the rat offspring. METHODS: Wistar rat dams were fed a control [normal-protein (NP); 17% protein] or an LP (8% protein) diet during pregnancy and lactation, and their male offspring were studied at 30 d of age. Direct measurements of baseline arterial blood pressure (ABP), heart rate (HR), and respiratory frequency (Rf) as well as peripheral chemoreflex activation (potassium cyanide: 0.04%) were recorded in pups while they were awake. In addition, recordings of the phrenic nerve (PN) and thoracic sympathetic nerve (tSN) activities were obtained from the in situ preparations. Hypoxia-inducible factor 1α (HIF-1α) expression was also evaluated in carotid bifurcation through a Western blotting assay. RESULTS: At 30 d of age, unanesthetized LP rats exhibited enhanced resting Rf (P = 0.001) and similar ABP and HR compared with the NP rats. Despite their similar baseline ABP values, LP rats exhibited augmented low-frequency variability (∼91%; P = 0.01). In addition, the unanesthetized LP rats showed enhanced pressor (P = 0.01) and tachypnoeic (P = 0.03) responses to peripheral chemoreflex activation. The LP rats displayed elevated baseline tSN activity (∼86%; P = 0.02) and PN burst frequency (45%; P = 0.01) and amplitude (53%; P = 0.001) as well as augmented sympathetic (P = 0.01) and phrenic (P = 0.04) excitatory responses to peripheral chemoreflex activation compared with the NP group. Furthermore, LP rats showed an increase of ∼100% in HIF-1α protein density in carotid bifurcation compared with NP rats. CONCLUSION: Sympathetic-respiratory overactivity and amplified peripheral chemoreceptor responses, potentially through HIF-1α-dependent mechanisms, precede the onset of hypertension in juvenile rats exposed to protein undernutrition during gestation and lactation.


Subject(s)
Chemoreceptor Cells/metabolism , Diet, Protein-Restricted/adverse effects , Maternal Nutritional Physiological Phenomena , Peripheral Nervous System/physiopathology , Prehypertension/physiopathology , Respiratory System/physiopathology , Sympathetic Nervous System/physiopathology , Animals , Birth Weight , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Carotid Artery, Common/physiopathology , Chemoreceptor Cells/pathology , Female , Fetal Development , Fetal Growth Retardation/etiology , Fetal Growth Retardation/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactation , Male , Peripheral Nervous System/pathology , Phrenic Nerve/pathology , Phrenic Nerve/physiopathology , Pregnancy , Prehypertension/etiology , Prehypertension/metabolism , Prehypertension/pathology , Rats, Wistar , Respiratory System/pathology , Sympathetic Nervous System/pathology , Thoracic Nerves/pathology , Thoracic Nerves/physiopathology
10.
Biomed Res Int ; 2015: 467597, 2015.
Article in English | MEDLINE | ID: mdl-26779536

ABSTRACT

Chronic heart failure (CHF) is a global health problem affecting millions of people. Autonomic dysfunction and disordered breathing patterns are commonly observed in patients with CHF, and both are strongly related to poor prognosis and high mortality risk. Tonic activation of carotid body (CB) chemoreceptors contributes to sympathoexcitation and disordered breathing patterns in experimental models of CHF. Recent studies show that ablation of the CB chemoreceptors improves autonomic function and breathing control in CHF and improves survival. These exciting findings indicate that alterations in CB function are critical to the progression of CHF. Therefore, better understanding of the physiology of the CB chemoreflex in CHF could lead to improvements in current treatments and clinical management of patients with CHF characterized by high chemosensitivity. Accordingly, the main focus of this brief review is to summarize current knowledge of CB chemoreflex function in different experimental models of CHF and to comment on their potential translation to treatment of human CHF.


Subject(s)
Carotid Body/physiopathology , Heart Failure/physiopathology , Respiration , Sympathetic Nervous System/physiopathology , Chemoreceptor Cells/pathology , Disease Progression , Humans , Reflex
11.
Proc Natl Acad Sci U S A ; 111(16): 6075-80, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24711432

ABSTRACT

Solitary chemosensory cells (SCCs) of the nasal cavity are specialized epithelial chemosensors that respond to irritants through the canonical taste transduction cascade involving Gα-gustducin and transient receptor potential melastatin 5. When stimulated, SCCs trigger peptidergic nociceptive (or pain) nerve fibers, causing an alteration of the respiratory rate indicative of trigeminal activation. Direct chemical excitation of trigeminal pain fibers by capsaicin evokes neurogenic inflammation in the surrounding epithelium. In the current study, we test whether activation of nasal SCCs can trigger similar local inflammatory responses, specifically mast cell degranulation and plasma leakage. The prototypical bitter compound, denatonium, a well-established activator of SCCs, caused significant inflammatory responses in WT mice but not mice with a genetic deletion of elements of the canonical taste transduction cascade, showing that activation of taste signaling components is sufficient to trigger local inflammation. Chemical ablation of peptidergic trigeminal fibers prevented the SCC-induced nasal inflammation, indicating that SCCs evoke inflammation only by neural activity and not by release of local inflammatory mediators. Additionally, blocking nicotinic, but not muscarinic, acetylcholine receptors prevents SCC-mediated neurogenic inflammation for both denatonium and the bacterial signaling molecule 3-oxo-C12-homoserine lactone, showing the necessity for cholinergic transmission. Finally, we show that the neurokinin 1 receptor for substance P is required for SCC-mediated inflammation, suggesting that release of substance P from nerve fibers triggers the inflammatory events. Taken together, these results show that SCCs use cholinergic neurotransmission to trigger peptidergic trigeminal nociceptors, which link SCCs to the neurogenic inflammatory pathway.


Subject(s)
Chemoreceptor Cells/pathology , Cholinergic Neurons/metabolism , Inflammation/pathology , Inflammation/physiopathology , Nose/pathology , Nose/physiopathology , Synaptic Transmission , Animals , Cell Degranulation , Chemoreceptor Cells/metabolism , Extravasation of Diagnostic and Therapeutic Materials/metabolism , Extravasation of Diagnostic and Therapeutic Materials/pathology , Extravasation of Diagnostic and Therapeutic Materials/physiopathology , Inflammation/metabolism , Mast Cells/physiology , Mice , Models, Biological , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Nasal Mucosa/physiopathology , Nociceptors/metabolism , Receptors, Neurokinin-1/metabolism , Receptors, Nicotinic/metabolism , Signal Transduction , TRPM Cation Channels/metabolism , Transducin/metabolism , Trigeminal Nerve/metabolism , Trigeminal Nerve/pathology
12.
Sheng Li Xue Bao ; 66(1): 23-9, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24553866

ABSTRACT

Peripheral chemoreceptors in the carotid body play important roles in the transduction of chemical stimuli in the arterial blood to the central for eliciting the chemoreflex, which mediates the ventilatory and circulatory responses to hypoxia. The activity of carotid chemoreceptor is modulated and significantly contributes to the ventilatory acclimatization at high altitude. In addition, the carotid chemoreceptor activity is augmented in patients with sleep-disordered breathing, notably in central or obstructive sleep apnea, and also in experimental animals. Thus, the carotid body functions to maintain the oxygen homeostasis, whereas anomalous carotid chemoreceptor activities could be both adaptive and pathogenic in sleep apnea. This review aims to summarize the cellular and molecular mechanisms that could mediate the augmented chemoreceptor activity induced by intermittent hypoxia. Our recent findings suggest a pathogenic role of inflammation mediated by an upregulation of renin-angiotensin system in the carotid body in the over-activity of the chemoreflex. These locally regulated mechanisms are proposed to be a significant part of the hypoxia-mediated maladaptive changes of the carotid body function, which could play a role in the pathophysiology of sleep apnea.


Subject(s)
Chemoreceptor Cells/pathology , Hypoxia/physiopathology , Sleep Apnea Syndromes/physiopathology , Acclimatization , Animals , Carotid Body/cytology , Humans , Renin-Angiotensin System
13.
Histochem Cell Biol ; 136(2): 191-204, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21688022

ABSTRACT

Epithelial tuft cells are named after their characteristic microtubule bundles located at the cell apex where these are exposed to the luminal environment. As such, tuft cells are found in multiple organs, including the gastrointestinal (GI) tract where the apical "tuft" is hypothesized to detect and transmit environmental signals. Thus, the goal of our study was to characterize gastric tuft cells during GI tract development, then subsequently in the normal and metaplastic adult stomach. GI tracts from mouse embryos, and newborn and postnatal mice were analyzed. Tuft cells were identified by immunohistochemistry using acetylated-α-tubulin (acTub) antibody to detect the microtubule bundle. Additional tuft cell markers, e.g., doublecortin-like kinase 1 (DCLK1), were used to co-localize with acTub. Tuft cells were quantified in human gastric tissue arrays and in mouse stomachs with or without inflammation. In the developing intestine, tuft cells in both the crypts and villi expressed all markers by E18.5. In the stomach, acTub co-localized with DCLK1 and other established tuft cell markers by E18.5 in the antrum, but not until postnatal day 7 in the corpus, with the highest density of tuft cells clustered at the forestomach ridge. Tuft cell numbers increased in hyperplastic human and mouse stomachs. In the adult GI tract, the tuft cell marker acTub co-expressed with DCKL1 and chemosensory markers, e.g.,TRPM5. In summary, tuft cells appear in the gastric antrum and intestine at E18.5, but their maximal numbers in the corpus are not achieved until after weaning. Tuft cell numbers increase with inflammation, hyperplasia, and metaplasia.


Subject(s)
Chemoreceptor Cells/metabolism , Chemoreceptor Cells/pathology , Gastric Mucosa/pathology , Protein Serine-Threonine Kinases/metabolism , Pyloric Antrum/pathology , Animals , Doublecortin-Like Kinases , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gastritis/metabolism , Gastritis/pathology , Gastrointestinal Tract/embryology , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/pathology , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Immunohistochemistry , Metaplasia/pathology , Mice , TRPM Cation Channels/metabolism
14.
Respir Physiol Neurobiol ; 178(3): 362-9, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21397054

ABSTRACT

Experiments in recent years have revealed labile electrophysiological and neurochemical phenotypes in primary afferent neurons exposed to specific stimulus conditions associated with the development of chronic pain. These studies collectively demonstrate that the mechanisms responsible for functional plasticity are primarily mediated by novel neuroimmune interactions involving circulating and resident immune cells and their secretory products, which together induce hyperexcitability in the primary sensory neurons. In another peripheral sensory modality, namely the arterial chemoreceptors, sustained stimulation in the form of chronic hypoxia (CH) elicits increased chemoafferent excitability from the mammalian carotid body. Previous studies which focused on functional changes in oxygen-sensitive type I cells in this organ have only partially elucidated the molecular and cellular mechanisms which initiate and control this adaptive response. Recent studies in our laboratory indicate a unique role for the immune system in regulating the chemo-adaptive response of the carotid body to physiologically relevant levels of hypoxia.


Subject(s)
Adaptation, Physiological , Carotid Body/pathology , Chemoreceptor Cells/metabolism , Chemoreceptor Cells/pathology , Chronic Pain/pathology , Animals , Carotid Body/physiology , Chemoreceptor Cells/physiology , Humans , Hypoxia/pathology , Inflammation/metabolism , Inflammation/pathology , Rats
15.
Neuroscience ; 175: 145-53, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21130843

ABSTRACT

Hypoxic respiratory and cardiovascular responses in mammals are mediated by peripheral chemoreceptor afferents which are relayed centrally via the solitary tract nucleus (NTS) in dorsomedial medulla to other cardiorespiratory-related brainstem regions such as ventrolateral medulla (VLM). Here, we test the hypothesis that peripheral chemoafferents could also be relayed directly to the Kölliker-Fuse/parabrachial complex in dorsolateral pons, an area traditionally thought to subserve pneumotaxic and cardiovascular regulation. Experiments were performed on adult Sprague-Dawley rats. Brainstem neurons with axons projecting to the dorsolateral pons were retrogradely labeled by microinjection with choleras toxin subunit B (CTB). Neurons involved in peripheral chemoreflex were identified by hypoxia-induced c-Fos expression. We found that double-labeled neurons (i.e. immunopositive to both CTB and c-Fos) were localized mostly in the commissural and medial subnuclei of NTS and to a lesser extent in the ventrolateral NTS subnucleus, VLM and ventrolateral pontine A5 region. Extracellular recordings from the commissural and medial NTS subnuclei revealed that some hypoxia-excited NTS neurons could be antidromically activated by electrical stimulations at the dorsolateral pons. These findings demonstrate that hypoxia-activated afferent inputs are relayed to the Kölliker-Fuse/parabrachial complex directly via the commissural and medial NTS and indirectly via the ventrolateral NTS subnucleus, VLM and A5 region. These pontine-projecting peripheral chemoafferent inputs may play an important role in the modulation of cardiorespiratory regulation by dorsolateral pons.


Subject(s)
Axons/pathology , Chemoreceptor Cells/pathology , Hypoxia/pathology , Pons/pathology , Respiratory Mechanics/physiology , Solitary Nucleus/pathology , Animals , Axons/physiology , Chemoreceptor Cells/physiology , Hypoxia/physiopathology , Male , Neural Pathways/pathology , Neural Pathways/physiology , Pons/physiology , Rats , Rats, Sprague-Dawley , Solitary Nucleus/physiology
16.
Respir Physiol Neurobiol ; 173(3): 322-35, 2010 Oct 31.
Article in English | MEDLINE | ID: mdl-20601214

ABSTRACT

The paired-like homeobox 2B gene (PHOX2B) is the disease-defining gene for congenital central hypoventilation syndrome (CCHS). Individuals with CCHS typically present in the newborn period with alveolar hypoventilation during sleep and often during wakefulness, altered respiratory control including reduced or absent ventilatory responses to hypercarbia and hypoxemia, and autonomic nervous system (ANS) dysregulation; however, a subset of individuals present well into adulthood. Thermoregulation is altered and perception of shortness of breath is absent, but voluntary breathing is retained. Structural and functional magnetic resonance imaging (MRI) and limited post-mortem studies in subjects with CCHS reveal abnormalities in both forebrain and brainstem. MRI changes appear in the hypothalamus (responsible for thermal drive to breathing), posterior thalamus and midbrain (mediating O(2) and oscillatory motor patterns), caudal raphé and locus coeruleus (regulating serotonergic and noradrenergic systems), the lateral medulla, parabrachial pons, and cerebellum (coordinating chemoreceptor and somatic afferent activity with breathing), and insular and cingulate cortices (mediating shortness of breath perception). Structural and functional alterations in these sites may result from PHOX2B mutations or be secondary to hypoxia/perfusion alterations from suboptimal management/compliance. The study of CCHS, with collaboration between physician-scientists and basic scientists, offers a rare opportunity to investigate control of breathing within the complex physiological network of the ANS.


Subject(s)
Autonomic Nervous System Diseases/genetics , Brain/pathology , Homeodomain Proteins/genetics , Hypoventilation/genetics , Respiratory Physiological Phenomena , Transcription Factors/genetics , Autonomic Nervous System/physiopathology , Autonomic Nervous System Diseases/congenital , Autonomic Nervous System Diseases/pathology , Chemoreceptor Cells/pathology , Humans , Hypoventilation/congenital , Hypoventilation/pathology , Infant, Newborn , Sleep Apnea, Central/congenital , Sleep Apnea, Central/genetics , Syndrome
17.
J Neurosci Res ; 88(12): 2755-64, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20544822

ABSTRACT

The aim of this study was to identify the effect of chemical activation of muscle metabosensitive afferent fibers from groups III and IV on Hoffmann (H-) reflex modulation in the vastus medialis muscle. The experiment was conducted in rats and was divided into two experiments. The first experiment consisted of recording the metabosensitive afferent activity from femoral nerve in rats in response to KCl intraarterial injections in nontreated adults and adults treated neonatally with capsaicin. Thus, the dose-response curve was determined. The second experiment consisted of eliciting the H- and M-waves before and after KCl injection in nontreated adult animals and those treated neonatally with capsaicin. Thus, the H(max)/M(max) ratio was measured. Results indicated that, 1) in nontreated animals, afferent fibers peak discharge was found after 10 mM KCl injection; 2) no significant increase in afferent discharge rate was found in capsaicin-treated animal after KCl injections, confirming that capsaicin is an excitotoxic agent that had destroyed the thin metabosensitive nerve fibers; 3) in nontreated animals, H(max)/M(max) ratio was significantly attenuated after a 10 mM KCl injection activating metabosensitive afferent fibers; and 4) in capsaicin-treated animals, no significant change in H(max)/M(max) ratio was observed after the KCl injection. These results reinforce the hypothesis that the spinal reflex response was influenced by metabosensitive muscle fibers and provide direct evidence that activation of these fibers could partially explain the reported decrease in H-reflex when metabolites are released in muscle.


Subject(s)
Chemoreceptor Cells/physiology , H-Reflex/physiology , Muscle, Skeletal/metabolism , Potassium/physiology , Sensory Receptor Cells/physiology , Spinal Cord/physiology , Action Potentials/drug effects , Action Potentials/physiology , Afferent Pathways/drug effects , Afferent Pathways/pathology , Afferent Pathways/physiology , Animals , Chemoreceptor Cells/drug effects , Chemoreceptor Cells/pathology , Female , Femoral Nerve/physiology , H-Reflex/drug effects , Male , Motor Neurons/physiology , Muscle, Skeletal/innervation , Quadriceps Muscle/innervation , Quadriceps Muscle/metabolism , Rats , Rats, Wistar , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology
18.
Proc Natl Acad Sci U S A ; 107(5): 2325-30, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20133877

ABSTRACT

Task2 K(+) channel expression in the central nervous system is surprisingly restricted to a few brainstem nuclei, including the retrotrapezoid (RTN) region. All Task2-positive RTN neurons were lost in mice bearing a Phox2b mutation that causes the human congenital central hypoventilation syndrome. In plethysmography, Task2(-/-) mice showed disturbed chemosensory function with hypersensitivity to low CO(2) concentrations, leading to hyperventilation. Task2 probably is needed to stabilize the membrane potential of chemoreceptive cells. In addition, Task2(-/-) mice lost the long-term hypoxia-induced respiratory decrease whereas the acute carotid-body-mediated increase was maintained. The lack of anoxia-induced respiratory depression in the isolated brainstem-spinal cord preparation suggested a central origin of the phenotype. Task2 activation by reactive oxygen species generated during hypoxia could silence RTN neurons, thus contributing to respiratory depression. These data identify Task2 as a determinant of central O(2) chemoreception and demonstrate that this phenomenon is due to the activity of a small number of neurons located at the ventral medullary surface.


Subject(s)
Carbon Dioxide/physiology , Oxygen/physiology , Potassium Channels, Tandem Pore Domain/physiology , Respiratory Center/physiology , Animals , Animals, Newborn , Brain Stem/pathology , Brain Stem/physiology , Brain Stem/physiopathology , Chemoreceptor Cells/pathology , Chemoreceptor Cells/physiology , Disease Models, Animal , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Humans , Hypercapnia/physiopathology , Hypoxia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Plethysmography, Whole Body , Potassium Channels, Tandem Pore Domain/deficiency , Potassium Channels, Tandem Pore Domain/genetics , Pregnancy , Respiratory Physiological Phenomena , Sleep Apnea, Central/etiology , Sleep Apnea, Central/genetics , Sleep Apnea, Central/physiopathology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology
19.
Int J Cardiol ; 142(1): 38-43, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-19176256

ABSTRACT

BACKGROUND: During the development of neurocardiogenic syncope (NCS) postural dependant venous blood pooling sets off a cascade of autonomic reflexes. This causes an initial rise in sympathetic tone, which is followed by an overshoot parasympathetic activation resulting in systemic vasodilatation and/or sinus bradycardia. However, other factors like associated hyperventilation or changes in blood gas content may also contribute to syncope. Hyperoxic cardiac chemoreflex sensitivity (CHRS) is an autonomic functional test that describes the heart rate decrease in response to increases in blood oxygen content. The purpose of this study was to investigate whether CHRS is altered in NCS. METHODS AND RESULTS: CHRS was compared in 16 NCS patients (49+/-4 yr old) vs. 16 age and gender matched controls (53+/-2 yr old). NCS was verified by clinical syncope and positive head-up tilt testing. The hyperoxic CHRS was measured by determination of the venous partial pressure of oxygen and heart rate before and after 5 min of pure oxygen inhalation. The difference of the R-R intervals before and after oxygen inhalation divided by the difference in the oxygen pressures were calculated as hyperoxic chemoreflex sensitivity [ms/mm Hg]. CHRS in the control group was 7.1+/-1.1 ms/mm Hg. By contrast, CHRS in NCS patients was significantly lower (2.8+/-1.0 ms/mm Hg; p<0.05). CONCLUSION: Neurocardiogenic syncope is associated with decreased hyperoxic cardiac chemoreflex sensitivity possibly reflecting impaired deactivation of arterial chemoreceptors. The clinical and pathophysiologic importance of chemosensor function in neurocardiogenic syncope needs to be investigated in more detail.


Subject(s)
Chemoreceptor Cells/physiology , Hyperoxia/diagnosis , Hyperoxia/physiopathology , Syncope, Vasovagal/diagnosis , Syncope, Vasovagal/physiopathology , Chemoreceptor Cells/pathology , Female , Humans , Hyperoxia/pathology , Male , Middle Aged , Syncope, Vasovagal/pathology , Tilt-Table Test/methods
20.
Dev Dyn ; 237(8): 2007-12, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18407555

ABSTRACT

The epithelial cells lining intrahepatic bile ducts (i.e., cholangiocytes), like many cell types in the body, have primary cilia extending from the apical plasma membrane into the bile ductal lumen. Cholangiocyte cilia express proteins such as polycystin-1, polycystin-2, fibrocystin, TRPV4, P2Y12, AC6, that account for ciliary mechano-, osmo-, and chemo-sensory functions; when these processes are disturbed by mutations in genes encoding ciliary-associated proteins, liver diseases (i.e., cholangiociliopathies) result. The cholangiociliopathies include but are not limited to cystic and fibrotic liver diseases associated with mutations in genes encoding polycystin-1, polycystin-2, and fibrocystin. In this review, we discuss the functions of cholangiocyte primary cilia, their role in the cholangiociliopathies, and potential therapeutic approaches.


Subject(s)
Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/pathology , Cilia/physiology , Epithelial Cells/ultrastructure , Liver Diseases/pathology , Animals , Chemoreceptor Cells/pathology , Chemoreceptor Cells/ultrastructure , Cilia/pathology , Epithelial Cells/pathology , Humans , Mechanoreceptors/pathology , Mechanoreceptors/ultrastructure , Water-Electrolyte Balance/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...