Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.588
Filter
1.
Physiol Rep ; 12(11): e16054, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872580

ABSTRACT

We aimed to determine the relative contribution of hypercapnia and hypoxia to the bradycardic response to apneas. We hypothesized that apneas with hypercapnia would cause greater bradycardia than normoxia, similar to the response seen with hypoxia, and that apneas with hypercapnic hypoxia would induce greater bradycardia than hypoxia or hypercapnia alone. Twenty-six healthy participants (12 females; 23 ± 2 years; BMI 24 ± 3 kg/m2) underwent three gas challenges: hypercapnia (+5 torr end tidal partial pressure of CO2 [PETCO2]), hypoxia (50 torr end tidal partial pressure of O2 [PETO2]), and hypercapnic hypoxia (combined hypercapnia and hypoxia), with each condition interspersed with normocapnic normoxia. Heart rate and rhythm, blood pressure, PETCO2, PETO2, and oxygen saturation were measured continuously. Hypercapnic hypoxic apneas induced larger bradycardia (-19 ± 16 bpm) than normocapnic normoxic apneas (-11 ± 15 bpm; p = 0.002), but had a comparable response to hypoxic (-19 ± 15 bpm; p = 0.999) and hypercapnic apneas (-14 ± 14 bpm; p = 0.059). Hypercapnic apneas were not different from normocapnic normoxic apneas (p = 0.134). After removal of the normocapnic normoxic heart rate response, the change in heart rate during hypercapnic hypoxia (-11 ± 16 bpm) was similar to the summed change during hypercapnia+hypoxia (-9 ± 10 bpm; p = 0.485). Only hypoxia contributed to this bradycardic response. Under apneic conditions, the cardiac response is driven by hypoxia.


Subject(s)
Apnea , Bradycardia , Heart Rate , Hypercapnia , Hypoxia , Humans , Hypercapnia/physiopathology , Female , Male , Heart Rate/physiology , Hypoxia/physiopathology , Apnea/physiopathology , Adult , Bradycardia/physiopathology , Young Adult , Blood Pressure/physiology , Carbon Dioxide/metabolism
2.
Int J Chron Obstruct Pulmon Dis ; 19: 1079-1091, 2024.
Article in English | MEDLINE | ID: mdl-38783895

ABSTRACT

Purpose: To develop and validate a nomogram for assessing the risk of developing hypercapnic respiratory failure (HRF) in patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD). Patients and Methods: From January 2019 to August 2023, a total of 334 AECOPD patients were enrolled in this research. We employed the Least Absolute Shrinkage and Selection Operator (LASSO) regression and multivariate logistic regression to determine independent predictors and develop a nomogram. This nomogram was appraised by the area under the receiver operating characteristic curve (AUC), calibration curve, Hosmer-Lemeshow goodness-of-fit test (HL test), decision curve analysis (DCA), and clinical impact curve (CIC). The enhanced bootstrap method was used for internal validation. Results: Sex, prognostic nutritional index (PNI), hematocrit (HCT), and activities of daily living (ADL) were independent predictors of HRF in AECOPD patients. The developed nomogram based on the above predictors showed good performance. The AUCs for the training, internal, and external validation cohorts were 0.841, 0.884, and 0.852, respectively. The calibration curves and HL test showed excellent concordance. The DCA and CIC showed excellent clinical usefulness. Finally, a dynamic nomogram was developed (https://a18895635453.shinyapps.io/dynnomapp/). Conclusion: This nomogram based on sex, PNI, HCT, and ADL demonstrated high accuracy and clinical value in predicting HRF. It is a less expensive and more accessible approach to assess the risk of developing HRF in AECOPD patients, which is more suitable for primary hospitals, especially in developing countries with high COPD-related morbidity and mortality.


Subject(s)
Disease Progression , Hypercapnia , Nomograms , Predictive Value of Tests , Pulmonary Disease, Chronic Obstructive , Respiratory Insufficiency , Humans , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Disease, Chronic Obstructive/complications , Male , Female , Aged , Hypercapnia/diagnosis , Hypercapnia/physiopathology , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/physiopathology , Respiratory Insufficiency/etiology , Risk Assessment , Risk Factors , Middle Aged , Reproducibility of Results , Prognosis , Nutrition Assessment , Aged, 80 and over , Hematocrit , Retrospective Studies , Sex Factors , Decision Support Techniques , Activities of Daily Living , Nutritional Status
3.
Nat Commun ; 15(1): 4475, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796568

ABSTRACT

About half of the neurons in the parabrachial nucleus (PB) that are activated by CO2 are located in the external lateral (el) subnucleus, express calcitonin gene-related peptide (CGRP), and cause forebrain arousal. We report here, in male mice, that most of the remaining CO2-responsive neurons in the adjacent central lateral (PBcl) and Kölliker-Fuse (KF) PB subnuclei express the transcription factor FoxP2 and many of these neurons project to respiratory sites in the medulla. PBclFoxP2 neurons show increased intracellular calcium during wakefulness and REM sleep and in response to elevated CO2 during NREM sleep. Photo-activation of the PBclFoxP2 neurons increases respiration, whereas either photo-inhibition of PBclFoxP2 or genetic deletion of PB/KFFoxP2 neurons reduces the respiratory response to CO2 stimulation without preventing awakening. Thus, augmenting the PBcl/KFFoxP2 response to CO2 in patients with sleep apnea in combination with inhibition of the PBelCGRP neurons may avoid hypoventilation and minimize EEG arousals.


Subject(s)
Carbon Dioxide , Forkhead Transcription Factors , Hypercapnia , Neurons , Parabrachial Nucleus , Wakefulness , Animals , Hypercapnia/physiopathology , Hypercapnia/metabolism , Neurons/metabolism , Neurons/physiology , Male , Parabrachial Nucleus/physiology , Parabrachial Nucleus/metabolism , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Mice , Carbon Dioxide/metabolism , Wakefulness/physiology , Respiration , Mice, Inbred C57BL , Calcitonin Gene-Related Peptide/metabolism , Sleep, REM/physiology , Repressor Proteins
4.
Sci Rep ; 14(1): 12262, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806563

ABSTRACT

Exercise elicits physiological adaptations, including hyperpnea. However, the mechanisms underlying exercise-induced hyperpnea remain unresolved. Skeletal muscle acts as a secretory organ, releasing irisin (IR) during exercise. Irisin can cross the blood-brain barrier, influencing muscle and tissue metabolism, as well as signaling in the central nervous system (CNS). We evaluated the effect of intracerebroventricular or intraperitoneal injection of IR in adult male rats on the cardiorespiratory and metabolic function during sleep-wake cycle under room air, hypercapnia and hypoxia. Central IR injection caused an inhibition on ventilation (VE) during wakefulness under normoxia, while peripheral IR reduced VE during sleep. Additionally, central IR exacerbates hypercapnic hyperventilation by increasing VE and reducing oxygen consumption. As to cardiovascular regulation, central IR caused an increase in heart rate (HR) across all conditions, while no change was observed following peripheral administration. Finally, central IR attenuated the hypoxia-induced regulated hypothermia and increase sleep episodes, while peripheral IR augmented CO2-induced hypothermia, during wakefulness. Overall, our results suggest that IR act mostly on CNS exerting an inhibitory effect on breathing under resting conditions, while stimulating the hypercapnic ventilatory response and increasing HR. Therefore, IR seems not to be responsible for the exercise-induced hyperpnea, but contributes to the increase in HR.


Subject(s)
Fibronectins , Physical Conditioning, Animal , Animals , Male , Rats , Fibronectins/metabolism , Hypercapnia/metabolism , Hypercapnia/physiopathology , Hypoxia/metabolism , Hypoxia/physiopathology , Heart Rate , Sleep/physiology , Wakefulness/physiology , Oxygen Consumption , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Respiration , Myokines
5.
Biosystems ; 241: 105231, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754621

ABSTRACT

OBJECTIVE: Dynamic cerebral autoregulation (dCA) has been addressed through different approaches for discriminating between normal and impaired conditions based on spontaneous fluctuations in arterial blood pressure (ABP) and cerebral blood flow (CF). This work presents a novel multi-objective optimisation (MO) approach for finding good configurations of a cerebrovascular resistance-compliance model. METHODS: Data from twenty-nine subjects under normo and hypercapnic (5% CO2 in air) conditions was used. Cerebrovascular resistance and vessel compliance models with ABP as input and CF velocity as output were fitted using a MO approach, considering fitting Pearson's correlation and error. RESULTS: MO approach finds better model configurations than the single-objective (SO) approach, especially for hypercapnic conditions. In addition, the Pareto-optimal front from the multi-objective approach enables new information on dCA, reflecting a higher contribution of myogenic mechanism for explaining dCA impairment.


Subject(s)
Cerebrovascular Circulation , Homeostasis , Humans , Cerebrovascular Circulation/physiology , Homeostasis/physiology , Linear Models , Male , Adult , Blood Pressure/physiology , Brain/physiology , Models, Cardiovascular , Hypercapnia/physiopathology , Female , Vascular Resistance/physiology
6.
Respir Physiol Neurobiol ; 326: 104269, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38688432

ABSTRACT

The neural control of breathing exhibits sex differences. There is now a large effort to account for biological sex in mammalian research, but the degree to which ectothermic vertebrates exhibit sex differences in the central control of breathing is not well-established. Therefore, we compared respiratory-related neural activity in brainstem-spinal cord preparations from female and male bullfrogs to determine if important aspects of the central control of breathing vary with sex. We found that the breathing pattern was similar across males and females, but baseline frequency of the respiratory network was faster in females. The magnitude of the central response to hypercapnia was similar across sexes, but the time to reach maximum burst rate occurred more slowly in females. These results suggest that sex differences may account for variation in traits associated with the control of breathing and that future work should carefully account for sex of the animal in analysis.


Subject(s)
Rana catesbeiana , Respiration , Sex Characteristics , Spinal Cord , Animals , Female , Male , Rana catesbeiana/physiology , Spinal Cord/physiology , Brain Stem/physiology , Hypercapnia/physiopathology
7.
Brain Struct Funct ; 229(5): 1121-1142, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38578351

ABSTRACT

In mammals, the ventral respiratory column (VRC) plays a pivotal role in integrating neurochemically diverse inputs from brainstem and forebrain regions to generate respiratory motor patterns. VRC microinjection of the neuropeptide galanin has been reported to dampen carbon dioxide (CO2)-mediated chemoreflex responses. Additionally, we previously demonstrated that galaninergic neurons in the retrotrapezoid nucleus (RTN) are implicated in the adaptive response to hypercapnic stimuli, suggesting a link between RTN neuroplasticity and increased neuronal drive to the VRC. VRC neurons express galanin receptor 1, suggesting potential regulatory action by galanin, however, the precise galaninergic chemoreceptor-VRC circuitry remains to be determined. This study aimed to identify sources of galaninergic input to the VRC that contribute to central respiratory chemoreception. We employed a combination of retrograde neuronal tracing, in situ hybridisation and immunohistochemistry to investigate VRC-projecting neurons that synthesise galanin mRNA. In an additional series of experiments, we used acute hypercapnia exposure (10% CO2, 1 h) and c-Fos immunohistochemistry to ascertain which galaninergic nuclei projecting to the VRC are activated. Our findings reveal that a total of 30 brain nuclei and 51 subnuclei project to the VRC, with 12 of these containing galaninergic neurons, including the RTN. Among these galaninergic populations, only a subset of the RTN neurons (approximately 55%) exhibited activation in response to acute hypercapnia. Our findings highlight that the RTN is the likely source of galaninergic transmission to the VRC in response to hypercapnic stimuli.


Subject(s)
Galanin , Hypercapnia , Neurons , Animals , Hypercapnia/metabolism , Hypercapnia/physiopathology , Male , Galanin/metabolism , Neurons/metabolism , Carbon Dioxide/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Neural Pathways/metabolism , Neural Pathways/physiology , Respiratory Center/metabolism , Rats , Chemoreceptor Cells/metabolism , Rats, Sprague-Dawley , Brain Stem/metabolism
8.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38673988

ABSTRACT

In Parkinson's disease (PD), along with typical motor dysfunction, abnormal breathing is present; the cause of which is not well understood. The study aimed to analyze the effects of stimulation of the serotonergic system with 5-HT1A and 5-HT2A agonists in a model of PD induced by injection of 6-hydroxydopamine (6-OHDA). To model PD, bilateral injection of 6-OHDA into both striata was performed in male Wistar rats. Respiratory disturbances in response to 7% hypercapnia (CO2 in O2) in the plethysmographic chamber before and after stimulation of the serotonergic system and the incidence of apnea were studied in awake rats 5 weeks after 6-OHDA or vehicle injection. Administration of 6-OHDA reduced the concentration of serotonin (5-HT), dopamine (DA) and norepinephrine (NA) in the striatum and the level of 5-HT in the brainstem of treated rats, which have been associated with decreased basal ventilation, impaired respiratory response to 7% CO2 and increased incidence of apnea compared to Sham-operated rats. Intraperitoneal (i.p.) injection of the 5-HT1AR agonist 8-OH-DPAT and 5-HT2AR agonist NBOH-2C-CN increased breathing during normocapnia and hypercapnia in both groups of rats. However, it restored reactivity to hypercapnia in 6-OHDA group to the level present in Sham rats. Another 5-HT2AR agonist TCB-2 was only effective in increasing normocapnic ventilation in 6-OHDA rats. Both the serotonergic agonists 8-OH-DPAT and NBOH-2C-CN had stronger stimulatory effects on respiration in PD rats, compensating for deficits in basal ventilation and hypercapnic respiration. We conclude that serotonergic stimulation may have a positive effect on respiratory impairments that occur in PD.


Subject(s)
Hypercapnia , Parkinson Disease , Receptor, Serotonin, 5-HT1A , Receptor, Serotonin, 5-HT2A , Animals , Male , Rats , Disease Models, Animal , Dopamine/metabolism , Hypercapnia/metabolism , Hypercapnia/physiopathology , Norepinephrine/metabolism , Norepinephrine/pharmacology , Oxidopamine/pharmacology , Parkinson Disease/metabolism , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Respiration/drug effects , Serotonin/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology
9.
Neurobiol Aging ; 139: 5-10, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38579393

ABSTRACT

Cerebrovascular reactivity (CVR) deficits may contribute to small vessel disease, such as white matter hyperintensities (WMH). Moreover, apolipoprotein-e4 (APOE4) carriers at genetic risk for Alzheimer's disease exhibit cerebrovascular dysfunction relative to non-carriers. We examined whether older adults, and APOE4 carriers specifically, with diminished CVR would exhibit higher WMH burden. Independently living older adults (N = 125, mean age = 69.2 years; SD = 7.6; 31.2% male) free of dementia or clinical stroke underwent brain MRI to quantify cerebral perfusion during CVR to hypercapnia and hypocapnia and determine WMH volume. Adjusting for age, sex and intracranial volume, hierarchical regression analysis revealed a significant association between whole brain CVR to hypercapnia and WMH overall [B = -.02, 95% CI (-.04, -.008), p =.003] and in APOE4 carriers [B = -.03, 95% CI (-.06, -.009), p =.009]. Findings suggest deficits in cerebral vasodilatory capacity are associated with WMH burden in older adults and future studies are warranted to further delineate the effect of APOE4 on precipitating WMH.


Subject(s)
Apolipoprotein E4 , Cerebrovascular Circulation , Magnetic Resonance Imaging , White Matter , Humans , Male , Female , Aged , White Matter/diagnostic imaging , White Matter/pathology , Apolipoprotein E4/genetics , Middle Aged , Aging/pathology , Aging/physiology , Heterozygote , Brain/diagnostic imaging , Brain/pathology , Brain/physiopathology , Brain/blood supply , Hypercapnia/physiopathology , Hypercapnia/diagnostic imaging , Risk , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/pathology
10.
Clin Auton Res ; 34(2): 297-301, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38502257

ABSTRACT

Increased sympathetic drive is of prognostic significance in chronic obstructive pulmonary disease (COPD) but its determinants remain poorly understood. One potential mechanism may be chemoreflex-mediated adrenergic stimulation caused by sustained hypercapnia. This study determined the impact of non-invasive ventilation (NIV) on muscle sympathetic nerve activity (MSNA) in patients with stable hypercapnic COPD. Ten patients (age 70 ± 7 years, GOLD stage 3-4) receiving long-term NIV (mean inspiratory positive airway pressure 21 ± 7 cmH2O) underwent invasive MSNA measurement via the peroneal nerve during spontaneous breathing and NIV. Compared with spontaneous breathing, NIV significantly reduced hypercapnia (PaCO2 51.5 ± 6.9 vs 42.6 ± 6.1 mmHg, p < 0.0001) along with the burst rate (64.4 ± 20.9 vs 59.2 ± 19.9 bursts/min, p = 0.03) and burst incidence (81.7 ± 29.3 vs 74.1 ± 26.9 bursts/100 heartbeats, p = 0.04) of MSNA. This shows for the first time that correcting hypercapnia with NIV decreases MSNA in COPD.


Subject(s)
Hypercapnia , Muscle, Skeletal , Noninvasive Ventilation , Pulmonary Disease, Chronic Obstructive , Sympathetic Nervous System , Humans , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Disease, Chronic Obstructive/therapy , Hypercapnia/therapy , Hypercapnia/physiopathology , Noninvasive Ventilation/methods , Male , Aged , Sympathetic Nervous System/physiopathology , Female , Middle Aged , Muscle, Skeletal/physiopathology , Muscle, Skeletal/innervation
11.
Respir Physiol Neurobiol ; 325: 104254, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38552704

ABSTRACT

We sought to determine if peripheral hypercapnic chemosensitivity is related to expiratory flow limitation (EFL) during exercise. Twenty participants completed one testing day which consisted of peripheral hypercapnic chemosensitivity testing and a maximal exercise test to exhaustion. The chemosensitivity testing consisting of two breaths of 10% CO2 (O2∼21%) repeated 5 times during seated rest and the first 2 exercise intensities during the maximal exercise test. Following chemosensitivity testing, participants continued cycling with the intensity increasing 20 W every 1.5 minutes till exhaustion. Maximal expiratory flow-volume curves were derived from forced expiratory capacity maneuvers performed before and after exercise at varying efforts. Inspiratory capacity maneuvers were performed during each exercise stage to determine EFL. There was no difference between the EFL and non-EFL hypercapnic chemoresponse (mean response during exercise 0.96 ± 0.46 and 0.91 ± 0.33 l min-1 mmHg-1, p=0.783). Peripheral hypercapnic chemosensitivity during mild exercise does not appear to be related to the development of EFL during exercise.


Subject(s)
Exercise Test , Exercise , Hypercapnia , Humans , Male , Hypercapnia/physiopathology , Exercise/physiology , Young Adult , Female , Adult , Tidal Volume/physiology , Tidal Volume/drug effects , Carbon Dioxide/metabolism
12.
J Exp Med ; 219(3)2022 03 07.
Article in English | MEDLINE | ID: mdl-35201268

ABSTRACT

Microglia, the main immunocompetent cells of the brain, regulate neuronal function, but their contribution to cerebral blood flow (CBF) regulation has remained elusive. Here, we identify microglia as important modulators of CBF both under physiological conditions and during hypoperfusion. Microglia establish direct, dynamic purinergic contacts with cells in the neurovascular unit that shape CBF in both mice and humans. Surprisingly, the absence of microglia or blockade of microglial P2Y12 receptor (P2Y12R) substantially impairs neurovascular coupling in mice, which is reiterated by chemogenetically induced microglial dysfunction associated with impaired ATP sensitivity. Hypercapnia induces rapid microglial calcium changes, P2Y12R-mediated formation of perivascular phylopodia, and microglial adenosine production, while depletion of microglia reduces brain pH and impairs hypercapnia-induced vasodilation. Microglial actions modulate vascular cyclic GMP levels but are partially independent of nitric oxide. Finally, microglial dysfunction markedly impairs P2Y12R-mediated cerebrovascular adaptation to common carotid artery occlusion resulting in hypoperfusion. Thus, our data reveal a previously unrecognized role for microglia in CBF regulation, with broad implications for common neurological diseases.


Subject(s)
Cerebrovascular Circulation/physiology , Microglia/physiology , Neurovascular Coupling/physiology , Receptors, Purinergic/physiology , Adult , Aged , Animals , Brain/physiology , Calcium Signaling/physiology , Carotid Artery Diseases/physiopathology , Evoked Potentials/physiology , Female , Humans , Hypercapnia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Purinergic P2Y12/physiology , Vasodilation/physiology , Vibrissae/innervation
13.
Sci Rep ; 12(1): 1099, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058534

ABSTRACT

In daily routine, many COPD patients report early onset augmented dyspnea following use of NIV (Deventilation Syndrome, DVS) as a negative side-effect. The aim of this study is the clinical characterization and concrete definition of DVS. This monocenter prospective observational study collected demographic, physiologic and symptomatic data from 67 in-patients with severe COPD Gold III-IV and chronic hypercapnic failure before, during and after use of an established NIV. During their inpatient follow-up, we examined patients during the first hour after termination of nocturnal NIV. DVS was defined by the authors as an increase of ≥ 2 points on the Borg scale during the first 30 min in patients who reported repeated dyspnea after the use of NIV. We monitored cardiovascular and respiratory data and measured diaphragm excursion. Subjective dyspnea was documented by use of the Borg scale and questionnaires. In addition, respirator and demographic data were collected. DVS occurred in 58% of our COPD patient collective, showing predominant emphysema phenotype. Patients with DVS were more severely ill than non-DVS concerning bronchial obstruction (FEV1 0.6 vs. 0.8 l, p < 0.05) and hypercapnia during spontaneous breathing (pre NIV pCO2: 54.5 vs. 49.3 mmHg, p < 0.02). DVS patients showed significantly higher respiratory rates (RR) (20.1 vs. 18.1/min p < 0.05) after termination of NIV. This trial characterizes and defines early onset augmented dyspnea after the use of NIV, referred to as DVS. It is hereby brought to attention as a frequent side effect of long-term home ventilation and possible pathophysiologic mechanisms are elucidated.


Subject(s)
Dyspnea/etiology , Noninvasive Ventilation/adverse effects , Pulmonary Disease, Chronic Obstructive/complications , Aged , Female , Humans , Hypercapnia/physiopathology , Lung/physiopathology , Male , Middle Aged , Positive-Pressure Respiration/adverse effects , Prospective Studies , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Disease, Chronic Obstructive/therapy , Respiration , Respiratory Insufficiency/etiology , Ventilators, Mechanical/adverse effects
14.
Respir Physiol Neurobiol ; 296: 103800, 2022 02.
Article in English | MEDLINE | ID: mdl-34626831

ABSTRACT

Infants born with neonatal opioid withdrawal syndrome (NOWS) can display abnormal cardiorespiratory patterns including tachypnea, tachycardia, and impaired ventilatory responses to hypoxia (HVR) and hypercapnia (HCVR). Chronic morphine exposure is associated with increased midbrain microglial expression. Using a rat model of pre- and post-natal morphine exposure, we assessed cardiorespiratory features of NOWS (resting tachycardia and tachypnea) including the attenuated HVR and HCVR and whether they are associated with increased brainstem microglia expression. Pregnant rats (dams) received twice-daily subcutaneous injections of morphine (5 mg/kg) during the third (last) week of pregnancy to simulate 3rd trimester in utero opioid exposure. Offspring then received once-daily subcutaneous injections of morphine (0.5 mg/kg) until postnatal (P) day P10 days of age to simulate postnatal morphine therapy. Cardiorespiratory responses were assessed 24 h later (P11 days) following spontaneous withdrawal. Compared to saline-treated pups, morphine-exposed offspring exhibited tachycardia and tachypnea as well as an attenuated HVR and HCVR. Microglial cell counts were increased in the nucleus tractus solitarius (nTS), dorsal motor nucleus of the vagus (DMNV) and nucleus ambiguous (NAamb), but not the retrapezoid nucleus (RTN) or the non-cardiorespriatory region, the cuneate nucleus (CN). These data suggest that the cardiorespiratory features and autonomic dysregulation in NOWS infants may be associated with altered microglial function in specific brainstem cardiorespiratory control regions.


Subject(s)
Brain Stem , Infant, Newborn, Diseases , Microglia , Opioid-Related Disorders , Substance Withdrawal Syndrome , Tachycardia , Tachypnea , Animals , Animals, Newborn , Brain Stem/immunology , Brain Stem/physiopathology , Disease Models, Animal , Female , Humans , Hypercapnia/immunology , Hypercapnia/physiopathology , Hypoxia/immunology , Hypoxia/physiopathology , Infant, Newborn , Infant, Newborn, Diseases/etiology , Infant, Newborn, Diseases/immunology , Infant, Newborn, Diseases/physiopathology , Microglia/immunology , Opioid-Related Disorders/complications , Opioid-Related Disorders/immunology , Opioid-Related Disorders/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Rats , Substance Withdrawal Syndrome/complications , Substance Withdrawal Syndrome/immunology , Substance Withdrawal Syndrome/physiopathology , Tachycardia/etiology , Tachycardia/immunology , Tachycardia/physiopathology , Tachypnea/etiology , Tachypnea/immunology , Tachypnea/physiopathology
15.
Respir Physiol Neurobiol ; 295: 103777, 2022 01.
Article in English | MEDLINE | ID: mdl-34425262

ABSTRACT

We reported that external dead space ventilation (EDSV) enhanced self-sustained muscle activity (SSMA) of the human soleus muscle, which is an indirect observation of plateau potentials. However, the main factor for EDSV to enhance SSMA remains unclear. The purpose of the present study was to examine the effects of EDSV-induced hypercapnia, hypoxia, and hyperventilation on SSMA. In Experiment 1 (n = 11; normal breathing [NB], EDSV, hypoxia, and voluntary hyperventilation conditions) and Experiment 2 (n = 9; NB and normoxic hypercapnia [NH] conditions), SSMA was evoked by electrical train stimulations of the right tibial nerve and measured using surface electromyography under each respiratory condition. In Experiment 1, SSMA was significantly higher than that in the NB condition only in the EDSV condition (P < 0.05). In Experiment 2, SSMA was higher in the NH condition than in the NB condition (P < 0.05). These results suggest that the EDSV-enhanced SSMA is due to hypercapnia, not hypoxia or increased ventilation.


Subject(s)
Chemoreceptor Cells/physiology , Hypercapnia/physiopathology , Hyperventilation/physiopathology , Hypoxia/physiopathology , Motor Neurons/physiology , Muscle, Skeletal/physiology , Adult , Electric Stimulation , Electromyography , Humans , Male
16.
Sci Rep ; 11(1): 23457, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34873185

ABSTRACT

Functional near infrared spectroscopy (fNIRS) measurements are confounded by signal components originating from multiple physiological causes, whose activities may vary temporally and spatially (across tissue layers, and regions of the cortex). Furthermore, the stimuli can induce evoked effects, which may lead to over or underestimation of the actual effect of interest. Here, we conducted a temporal, spectral, and spatial analysis of fNIRS signals collected during cognitive and hypercapnic stimuli to characterize effects of functional versus systemic responses. We utilized wavelet analysis to discriminate physiological causes and employed long and short source-detector separation (SDS) channels to differentiate tissue layers. Multi-channel measures were analyzed further to distinguish hemispheric differences. The results highlight cardiac, respiratory, myogenic, and very low frequency (VLF) activities within fNIRS signals. Regardless of stimuli, activity within the VLF band had the largest contribution to the overall signal. The systemic activities dominated the measurements from the short SDS channels during cognitive stimulus, but not hypercapnic stimulus. Importantly, results indicate that characteristics of fNIRS signals vary with type of the stimuli administered as cognitive stimulus elicited variable responses between hemispheres in VLF band and task-evoked temporal effect in VLF, myogenic and respiratory bands, while hypercapnic stimulus induced a global response across both hemispheres.


Subject(s)
Brain Mapping/methods , Brain/physiology , Cerebral Cortex/physiology , Cognition/physiology , Hypercapnia/physiopathology , Spectroscopy, Near-Infrared/methods , Adult , Biomarkers/metabolism , Female , Humans , Male , Neurosciences , Principal Component Analysis , Statistics as Topic , Wavelet Analysis , Young Adult
17.
Dtsch Med Wochenschr ; 146(22): 1497-1508, 2021 11.
Article in German | MEDLINE | ID: mdl-34741295

ABSTRACT

COPD is the most common reason for hypercapnia. However, it is -by far- not the only reason. In fact, numerous neuromuscular disorders (not only ALS) as well as restrictive thoracic disorders do also lead to clinically highly relevant hypercapnia. Early diagnosis of hypercapnic ventilatory failure usually takes place at nighttime. NIV devices work with a periodic interplay of alternating IPAP and EPAP which results in a ventilation of the lungs, thereby elimination CO2 to treat hypercapnic respiratory failure. Firstline settings for a NIV therapy to treat "stable hypercapnia" are as follows: Pressure Support Ventilation Modus, EPAP 5 cmH2O, IPAP 15 cmH2O, Back Up rate 15/Minute. The overall goal of NIV treatment is a successful reduction in CO2. This can be achieved by changing the following variables of the ventilator settings: increase in IPAP ± increase in back up respiratory rate ± use of assisted pressure controlled ventilation mode (APCV).


Subject(s)
Home Care Services , Hypercapnia , Noninvasive Ventilation , Aged , Humans , Hypercapnia/physiopathology , Hypercapnia/therapy , Neuromuscular Diseases/therapy , Pulmonary Disease, Chronic Obstructive/therapy
18.
Sci Rep ; 11(1): 20557, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663876

ABSTRACT

The roles of endothelial nitric oxide synthase (eNOS) in the ventilatory responses during and after a hypercapnic gas challenge (HCC, 5% CO2, 21% O2, 74% N2) were assessed in freely-moving female and male wild-type (WT) C57BL6 mice and eNOS knock-out (eNOS-/-) mice of C57BL6 background using whole body plethysmography. HCC elicited an array of ventilatory responses that were similar in male and female WT mice, such as increases in breathing frequency (with falls in inspiratory and expiratory times), and increases in tidal volume, minute ventilation, peak inspiratory and expiratory flows, and inspiratory and expiratory drives. eNOS-/- male mice had smaller increases in minute ventilation, peak inspiratory flow and inspiratory drive, and smaller decreases in inspiratory time than WT males. Ventilatory responses in female eNOS-/- mice were similar to those in female WT mice. The ventilatory excitatory phase upon return to room-air was similar in both male and female WT mice. However, the post-HCC increases in frequency of breathing (with decreases in inspiratory times), and increases in tidal volume, minute ventilation, inspiratory drive (i.e., tidal volume/inspiratory time) and expiratory drive (i.e., tidal volume/expiratory time), and peak inspiratory and expiratory flows in male eNOS-/- mice were smaller than in male WT mice. In contrast, the post-HCC responses in female eNOS-/- mice were equal to those of the female WT mice. These findings provide the first evidence that the loss of eNOS affects the ventilatory responses during and after HCC in male C57BL6 mice, whereas female C57BL6 mice can compensate for the loss of eNOS, at least in respect to triggering ventilatory responses to HCC.


Subject(s)
Nitric Oxide Synthase Type III/metabolism , Pulmonary Ventilation/genetics , Pulmonary Ventilation/physiology , Animals , Female , Hypercapnia/physiopathology , Hypoxia , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/physiology , Respiration , Respiratory Insufficiency/physiopathology , Tidal Volume
19.
Physiol Rep ; 9(20): e15035, 2021 10.
Article in English | MEDLINE | ID: mdl-34665531

ABSTRACT

PURPOSE: As part of our investigations of intraocular pressure (IOP) as a potential contributing factor to the spaceflight-associated neuro-ocular syndrome using the 6° head-down tilt (6°HDT) bed rest experimental model, we compared the effect of rest and isometric exercise in prone and supine 6°HDT positions on IOP with that observed in the seated position. METHODS: Ten male volunteers (age = 22.5 ± 3.1 yrs) participated in six interventions. All trials comprised a 10-min rest period, a 3-min isometric handgrip exercise at 30% of participant's maximum, and a 10-min recovery period. The trials were conducted under normocapnic (NCAP) or hypercapnic (FI CO2  = 0.01; HCAP) conditions, the latter mimicking the ambient conditions on the International Space Station. IOP, systolic and diastolic pressures, and heart rate (HR) were measured during the trials. RESULTS: Isometric exercise-induced elevations in HR and mean arterial blood pressure. IOP in the prone 6°HDT position was significantly higher (p < 0.001) compared to IOP in supine 6°HDT position and seated trials at all time points. IOP increased with exercise only in a seated HCAP trial (p = 0.042). No difference was observed between trials in NCAP and HCAP. IOP in the prone 6°HDT position was constantly elevated above 21 mmHg, the lower limit for clinical ocular hypertension. CONCLUSIONS: IOP in the prone 6°HDT position was similar to IOP reported in astronauts upon entering microgravity, potentially indicating that prone, rather than supine 6°HDT position might be a more suitable experimental analog for investigating the acute ocular changes that occur in microgravity.


Subject(s)
Exercise , Hand Strength , Hypercapnia/physiopathology , Intraocular Pressure , Prone Position , Supine Position , Adult , Bed Rest , Head-Down Tilt , Heart Rate , Humans , Male , Weightlessness Simulation , Young Adult
20.
Physiol Rep ; 9(19): e15021, 2021 10.
Article in English | MEDLINE | ID: mdl-34617685

ABSTRACT

Squat-stand maneuvers (SSMs) are a popular method of inducing blood pressure (BP) oscillations to reliably assess dynamic cerebral autoregulation (dCA), but their effects on the cerebral circulation remain controversial. We designed a protocol whereby participants would perform SSMs under hypercapnic conditions. Alarmingly high values of cerebral blood flow velocity (CBFV) were recorded, leading to early study termination after the recruitment of a single participant. One healthy subject underwent recordings at rest (5 min sitting, 5 min standing) and during two SSMs (fixed and random frequency). Two sets of recordings were collected; one while breathing room air, one while breathing 5% CO2 . Continuous recordings of bilateral CBFV (transcranial Doppler), heart rate (ECG), BP (Finometer), and end-tidal CO2 (capnography) were collected. Peak values of systolic CBFV were significantly higher during hypercapnia (p < 0.01), and maximal values exceeded 200 cm.s-1 . Estimates of dCA (ARI) during hypercapnia were impaired relative to poikilocapnia (p = 0.03). The phase was significantly reduced under hypercapnic conditions (p = 0.03). Here we report extremely high values of CBFV in response to repeated SSMs during induced hypercapnia, in an otherwise healthy subject. Our findings suggest that protocols performing hypercapnic SSMs are potentially dangerous. We, therefore, urge caution if other research groups plan to undertake similar protocols.


Subject(s)
Blood Pressure/physiology , Cerebrovascular Circulation/physiology , Exercise/physiology , Heart Rate/physiology , Hypercapnia/physiopathology , Blood Flow Velocity/physiology , Electroencephalography , Homeostasis/physiology , Humans , Male , Posture/physiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...