Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.019
Filter
1.
Biochem Pharmacol ; 195: 114805, 2022 01.
Article in English | MEDLINE | ID: mdl-34673011

ABSTRACT

Opioid-related fatalities involving synthetic opioids have reached unprecedented levels. This study evaluated the respiratory depressant effects of seven fentanyl analogs that have either emerged in the illicit drug supply or been identified in toxicological analyses following fatal or non-fatal intoxications. Adult male Swiss Webster mice were administered fentanyl analogs (isobutyrylfentanyl, crotonylfentanyl, para-methoxyfentanyl, para-methoxybutyrylfentanyl, 3-furanylfentanyl, thiophenefentanyl, and benzodioxolefentanyl) and their effects on minute volume as compared to mu-opioid receptor (MOR) agonist standards (fentanyl, morphine, and buprenorphine) were measured using whole body plethysmography (WBP). All drugs elicited significant (p ≤ 0.05) hypoventilation relative to vehicle for at least one dose tested: morphine (1, 3.2, 10, 32 mg/kg), buprenorphine, (0.032, 0.1, 0.32, 1, 3.2 mg/kg), fentanyl (0.0032, 0.01, 0.032, 0.1, 1, 32 mg/kg), isobutyrylfentanyl (0.1, 0.32, 1, 3.2, 10 mg/kg), crotonylfentanyl (0.1, 0.32, 1, 3.2, 10 mg/kg), para-methoxyfentanyl (0.1, 0.32, 1, 3.2, 10 mg/kg), para-methoxybutyrylfentanyl (0.32, 1, 3.2, 10 mg/kg), 3-furanylfentanyl (0.1, 0.32, 1, 3.2, 10 mg/kg), thiophenefentanyl (1, 3.2, 10, 32, 100 mg/kg), and benzodioxolefentanyl (3.2, 10, 32, 100 mg/kg). The ED50 values for hypoventilation showed a rank order of potency as follows: fentanyl (ED50 = 0.96 mg/kg) > 3-furanylfentanyl (ED50 = 2.60 mg/kg) > crotonylfentanyl (ED50 = 2.72 mg/kg) > para-methoxyfentanyl (ED50 = 3.31 mg/kg) > buprenorphine (ED50 = 10.8 mg/kg) > isobutyrylfentanyl (ED50 = 13.5 mg/kg) > para-methoxybutyrylfentanyl (ED50 = 16.1 mg/kg) > thiophenefentanyl (ED50 = 18.0 mg/kg) > morphine (ED50 = 55.3 mg/kg) > benzodioxolefentanyl (ED50 = 10,168 mg/kg). A naloxone pretreatment (10 mg/kg) attenuated the hypoventilatory effects of all drugs. These results establish that the respiratory depressant effects of these fentanyl analogs are at least in part mediated by the MOR.


Subject(s)
Fentanyl/analogs & derivatives , Hypoventilation/prevention & control , Naloxone/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Animals , Fentanyl/chemistry , Fentanyl/toxicity , Hypoventilation/chemically induced , Hypoventilation/physiopathology , Male , Mice , Molecular Structure , Narcotic Antagonists/pharmacology , Plethysmography/methods , Receptors, Opioid, mu/physiology , Respiratory Mechanics/drug effects , Respiratory Mechanics/physiology
2.
Respir Physiol Neurobiol ; 294: 103768, 2021 12.
Article in English | MEDLINE | ID: mdl-34343692

ABSTRACT

Acute intermittent hypoxia (AIH) modifies the functioning of the respiratory network, causing respiratory motor facilitation in anesthetized animals and a compensatory increase in pulmonary ventilation in freely behaving animals. However, it is still unclear whether the ventilatory facilitation induced by AIH in unanesthetized animals is associated with changes in the respiratory pattern. We found that Holtzman male rats (80-150 g) exposed to AIH (10 × 6% O2 for 30-40 s every 5 min, n = 9) exhibited a prolonged (30 min) increase in baseline minute ventilation (P < 0.05) compared to control animals (n = 13), combined with the occurrence of late expiratory peak flow events, suggesting the presence of active expiration. The increase in ventilation after AIH was also accompanied by reductions in arterial CO2 and body temperature (n = 5-6, P < 0.05). The systemic treatment with ketanserin (a 5-HT2 receptor antagonist) before AIH prevented the changes in ventilation and active expiration (n = 11) but potentiated the hypothermic response (n = 5, P < 0.05) when compared to appropriate control rats (n = 13). Our findings indicate that the ventilatory long-term facilitation elicited by AIH exposure in unanesthetized rats is linked to the generation of active expiration by mechanisms that may depend on the activation of serotonin receptors. In contrast, the decrease in body temperature induced by AIH may not require 5-HT2 receptor activation.


Subject(s)
Hypoxia/physiopathology , Ketanserin/pharmacology , Pulmonary Ventilation/physiology , Respiratory Mechanics/physiology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Disease Models, Animal , Male , Pulmonary Ventilation/drug effects , Rats , Rats, Sprague-Dawley , Respiratory Mechanics/drug effects , Tidal Volume/physiology
3.
Am J Physiol Regul Integr Comp Physiol ; 321(4): R558-R571, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34405704

ABSTRACT

Orexin neurons are active in wakefulness and mostly silent in sleep. In adult rats and humans, orexin facilitates the hypercapnic ventilatory response but has little effect on resting ventilation. The influence of orexin on breathing in the early postnatal period, and across states of vigilance, have not been investigated. This is relevant as the orexin system may be impaired in Sudden Infant Death Syndrome (SIDS) cases. We addressed three hypotheses: 1) orexin provides a drive to breathe in infancy; 2) the effect of orexin depends on stage of postnatal development; and 3) orexin has a greater influence on breathing in wakefulness compared with sleep. Whole body plethysmography was used to monitor breathing of infant rats at three ages: postnatal days (P) 7-8, 12-14, and 17-19. Respiratory variables were analyzed in wakefulness (W), quiet sleep (QS), and active sleep (AS), following suvorexant (5 mg/kg ip), a dual orexin receptor antagonist, or vehicle (DMSO). Effects of suvorexant on ventilatory responses to graded hypercapnia ([Formula: see text] = 0.02, 0.04, 0.06), hypoxia ([Formula: see text] = 0.10), and hyperoxia ([Formula: see text] = 1.0) at P12-14 were also tested. At P12-14, but not at other ages, suvorexant significantly reduced respiratory frequency in all states, reduced the ventilatory equivalent in QW and QS, and increased [Formula: see text] to ∼5 mmHg. Suvorexant had no effect on ventilatory responses to graded hypercapnia or hypoxia. Hyperoxia eliminated the effects of suvorexant on respiratory frequency at P12-14. Our data suggest that orexin preserves eupneic frequency and ventilation in rats, specifically at ∼2 wk of age, perhaps by facilitating tonic peripheral chemoreflex activity.


Subject(s)
Chemoreceptor Cells/metabolism , Lung/innervation , Orexins/metabolism , Pulmonary Ventilation , Reflex , Respiratory Mechanics , Animals , Animals, Newborn , Azepines/pharmacology , Chemoreceptor Cells/drug effects , Hypercapnia/metabolism , Hypercapnia/physiopathology , Hypoxia/metabolism , Hypoxia/physiopathology , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/metabolism , Pulmonary Ventilation/drug effects , Rats, Sprague-Dawley , Reflex/drug effects , Respiratory Mechanics/drug effects , Sleep , Triazoles/pharmacology , Wakefulness
4.
Exp Neurol ; 345: 113813, 2021 11.
Article in English | MEDLINE | ID: mdl-34284029

ABSTRACT

Thyroid hormones (THs) are essential for foetal brain development. Because the gestating mother is the main source of THs to the foetus, maternal hypothyroidism and/or premature birth compromise neurological outcomes in the offspring. Respiratory instability and recurrent apneas due to immaturity of the respiratory control network are major causes of morbidity in infants. Inadequate TH supply may be sufficient to delay perinatal maturation of the respiratory control system; however, this hypothesis remains untested. To address this issue, maternal hypothyroidism was induced by adding methimazole (MMI; 0.02% w/v) to the drinking water of pregnant dams from conception to postpartum day 4 (P4). The effect of TH supplementation on respiratory function was tested by injecting levothyroxine (L-T4) in newborns at P1. Respiratory function was assessed by plethysmography (in vivo) and recording of phrenic output from medullary preparations (in vitro). By comparison with controls, TH deficiency increased the frequency of apneas and decreased basal ventilation in vivo and prevented the age-dependent increase in phrenic burst frequency normally observed in vitro. The effects of TH deficiency on GABAergic modulation of respiratory activity were measured by bath application of muscimol (GABAA agonist) or bicuculline (GABAA antagonist). The phrenic burst frequency responses to GABAergic agents were consistently greater in preparations from TH deficient pups. L-T4 supplementation reversed part of the respiratory anomalies related to MMI treatment in vitro. We conclude that TH deficiency during the perinatal period is sufficient to delay maturation of the respiratory control network development. Excessive GABAergic inhibition may contribute to this effect.


Subject(s)
Antithyroid Agents/pharmacology , Nerve Net/metabolism , Phrenic Nerve/metabolism , Respiratory Mechanics/physiology , Thyroid Hormones/deficiency , Animals , Animals, Newborn , Female , GABA-A Receptor Antagonists/pharmacology , Male , Methimazole/pharmacology , Nerve Net/drug effects , Phrenic Nerve/drug effects , Plethysmography/methods , Pregnancy , Rats , Rats, Sprague-Dawley , Respiration/drug effects , Respiratory Mechanics/drug effects
5.
Toxicol Appl Pharmacol ; 427: 115652, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34298059

ABSTRACT

Exposure to phosphine (PH3) presents with a host of diverse, non-specific symptoms that span multiple organ systems and is characterized by a high mortality rate. While a comprehensive mechanism for PH3 poisoning remains inconclusive, prior studies have implicated cardiac failure and circulatory compromise as potential pathways central to PH3-induced mortality. In this study, milrinone (MLR), a phosphodiesterase-3 inhibitor used to treat cardiac failure, was investigated as a potential countermeasure for PH3 poisoning. Lethality, physiological responses, and behavioral changes were evaluated in telemetrized female rats pretreated with water (sham) or one of three doses of MLR (40, 200, or 600 µg/kg) and exposed to PH3 (660 ppm for 25-40 min; 16,500-26,400 ppm × min). Animals receiving prophylactic administration of 600 µg/kg of MLR had nominally improved survivability compared to sham animals, although median lethal concentration-time and time of death did not differ substantially between treatment groups. Changes in respiration and behavior induced by PH3 appeared largely unaffected by MLR pretreatment, regardless of dose. Conversely, MLR pretreatment alleviated some aspects of PH3-induced cardiac function impairment, with slight dose-dependent effects observed for cardiac contractility, mean arterial pressure, and QRS duration. Together, these results illustrate the importance of circulatory compromise in PH3 poisoning and highlight the potential viability of MLR as a potential countermeasure option or part of a countermeasure regimen when administered prophylactically at 600 µg/kg.


Subject(s)
Cardiac Output/drug effects , Cardiotonic Agents/administration & dosage , Insecticides/poisoning , Milrinone/administration & dosage , Phosphines/poisoning , Respiratory Mechanics/drug effects , Animals , Cardiac Output/physiology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Inhalation Exposure/adverse effects , Lethal Dose 50 , Pre-Exposure Prophylaxis/methods , Rats , Rats, Sprague-Dawley , Respiratory Mechanics/physiology , Survival Rate/trends
6.
Respir Physiol Neurobiol ; 292: 103705, 2021 10.
Article in English | MEDLINE | ID: mdl-34062282

ABSTRACT

Balb/c mice respiratory mechanics was studied in two intravenous methacholine (MCh) protocols: bolus and continuous infusion. The Constant Phase Model (CPM) was used in this study. The harmonic distortion index (kd) was used to assess the respiratory system nonlinearity. The analysis of variance showed difference between groups (OVA vs control) and among doses for both protocols. Bolus protocol posttest: there was a difference between OVA and control at 0.3 and 1 mg/kg doses (p<0.0001 and p<0.001) for Rn. Infusion: there was a difference between OVA and control at 192 µg.kg-1.min-1 dose for Rn, G and H, (p<0.01; p<0.001; p<0.001). An increment was found in kd values near to the observed peak values in bolus protocol. The bolus protocol could better differentiate inflamed and non-inflamed airway resistance, whereas the differences between OVA and control in continuous infusion protocol were associated to airway- and, mainly, parenchyma-related parameters. Moreover, the bolus protocol presented a higher nonlinear degree compared to the infusion protocol.


Subject(s)
Asthma/chemically induced , Bronchoconstrictor Agents/administration & dosage , Methacholine Chloride/administration & dosage , Models, Theoretical , Respiratory Mechanics/drug effects , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C
7.
Acta Biochim Biophys Sin (Shanghai) ; 53(8): 1076-1087, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34137445

ABSTRACT

Propofol is the most commonly used intravenous anesthetic worldwide. It can induce loss of consciousness prior to the occurrence of severe respiratory suppression, which is also a pharmacodynamic feature of all general anesthetics. However, the neural mechanisms underlying this natural phenomenon are controversial and highly related to patient safety. In the present study, we demonstrated that the pharmacodynamic effects of propofol (50 and 100 µM) on suppression of consciousness-related excitatory postsynaptic currents in the medial prefrontal cortex (mPFC) and centromedian nucleus of the thalamus (CMT) were lower than those in the kernel respiratory rhythmogenesis nucleus pre-Bötzinger complex (PrBo). Furthermore, we unexpectedly found that the GABAA receptor ß3 subunit is the key target for propofol's action and that it is mutually and exclusively expressed in GABAergic neurons. It is also more abundant in the mPFC and CMT, but mainly co-localized with GABAergic neurons in the PrBo. As a result, the differentiated expression pattern should mediate more neuron suppression through the activation of GABAergic neurons in the mPFC and CMT at low doses of propofol (50 µM). However, PrBo GABAergic neurons were only activated by propofol at a high dose (100 µM). These results highlight the detailed pharmacodynamic effects of propofol on consciousness-related and respiration-related nuclei and provide the distinct interaction mechanism between the ß3 subunit and GABAergic neurons in mediating the suppression of consciousness compared to the inhibition of respiration.


Subject(s)
GABAergic Neurons/metabolism , Intralaminar Thalamic Nuclei , Prefrontal Cortex , Propofol/pharmacology , Receptors, GABA-A/metabolism , Respiratory Mechanics/drug effects , Unconsciousness , Animals , Intralaminar Thalamic Nuclei/metabolism , Intralaminar Thalamic Nuclei/physiopathology , Male , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Rats , Rats, Sprague-Dawley , Unconsciousness/chemically induced , Unconsciousness/metabolism , Unconsciousness/physiopathology
8.
Crit Care Med ; 49(12): 2090-2101, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34115638

ABSTRACT

OBJECTIVES: In mechanically ventilated patients, deep sedation is often assumed to induce "respirolysis," that is, lyse spontaneous respiratory effort, whereas light sedation is often assumed to preserve spontaneous effort. This study was conducted to determine validity of these common assumptions, evaluating the association of respiratory drive with sedation depth and ventilator-free days in acute respiratory failure. DESIGN: Prospective cohort study. SETTING: Patients were enrolled during 2 month-long periods in 2016-2017 from five ICUs representing medical, surgical, and cardiac specialties at a U.S. academic hospital. PATIENTS: Eligible patients were critically ill adults receiving invasive ventilation initiated no more than 36 hours before enrollment. Patients with neuromuscular disease compromising respiratory function or expiratory flow limitation were excluded. INTERVENTIONS: Respiratory drive was measured via P0.1, the change in airway pressure during a 0.1-second airway occlusion at initiation of patient inspiratory effort, every 12 ± 3 hours for 3 days. Sedation depth was evaluated via the Richmond Agitation-Sedation Scale. Analyses evaluated the association of P0.1 with Richmond Agitation-Sedation Scale (primary outcome) and ventilator-free days. MEASUREMENTS AND MAIN RESULTS: Fifty-six patients undergoing 197 bedside evaluations across five ICUs were included. P0.1 ranged between 0 and 13.3 cm H2O (median [interquartile range], 0.1 cm H2O [0.0-1.3 cm H2O]). P0.1 was not significantly correlated with the Richmond Agitation-Sedation Scale (RSpearman, 0.02; 95% CI, -0.12 to 0.16; p = 0.80). Considering P0.1 terciles (range less than 0.2, 0.2-1.0, and greater than 1.0 cm H2O), patients in the middle tercile had significantly more ventilator-free days than the lowest tercile (incidence rate ratio, 0.78; 95% CI, 0.65-0.93; p < 0.01) or highest tercile (incidence rate ratio, 0.58; 95% CI, 0.48-0.70; p < 0.01). CONCLUSIONS: Sedation depth is not a reliable marker of respiratory drive during critical illness. Respiratory drive can be low, moderate, or high across the range of routinely targeted sedation depth.


Subject(s)
Hypnotics and Sedatives/classification , Respiratory Mechanics/drug effects , Adult , Aged , Cohort Studies , Critical Illness/therapy , Female , Humans , Hypnotics and Sedatives/adverse effects , Male , Middle Aged , Ontario , Prospective Studies , Respiration, Artificial/methods , Respiration, Artificial/statistics & numerical data , Respiratory Mechanics/physiology , Retrospective Studies
9.
J Neurophysiol ; 125(5): 1543-1551, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33760672

ABSTRACT

Eupnea and gasping in infancy depend on central nervous system (CNS) serotonin (5-hydroxytryptamine; 5-HT). Although previous in vitro preparations have provided some evidence that 5-HT acts through type 2 A receptors (5-HT2A) to facilitate eupnea and gasping, here the hypothesis addressed is that 5-HT2A receptor activation is necessary for eupnea and the proper generation of gasping in vivo. To test this, we administered 2,5-dimethoxy-4-iodoamphetamine (DOI; 0.25 mg/kg i.p.), a 5-HT2A agonist, 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT; 0.25 mg/kg i.p.), a 5-HT1A agonist, or vehicle (saline) to 7-9-day-old tryptophan hydroxylase 2 knockout (TPH2-/-) mice. A second experiment assessed the effect of MDL-11,939 (MDL; 10 mg/kg i.p.), the specific 5-HT2A antagonist, or vehicle (DMSO) on the gasping of wild-type (TPH2+/+) animals. Drugs were given 15 min prior to five episodes of severe hypoxia that elicited gasping. TPH2-/- breathed more slowly but had the same V̇e and V̇e/V̇o2 compared with TPH2+/+. As previously reported, the gasping of TPH2-/- was significantly delayed (P < 0.001) and occurred at a significantly lower frequency compared with TPH2+/+ (P = 0.04). For both genotypes, DOI hastened eupneic frequency but had no effect on V̇e or V̇e/V̇o2. The gasping of TPH2-/-, although unaffected by 8-OH-DPAT, was indistinguishable from the gasping of TPH2+/+ following DOI. In TPH2+/+, application of MDL led to hypoventilation (P = 0.01), a delay in the appearance of gasping (P = 0.005), and reduced gasp frequency (P = 0.05). These data show that, in vivo, 5-HT2A receptors facilitate both eupnea and gasping. As has been shown in vitro, 5-HT2A probably promotes gasping by exciting hypoxia-resistant pacemaker neurons.NEW & NOTEWORTHY Previous in vitro studies suggest that 5-HT2A receptors contribute to eupnea and are necessary for fictive gasping. The current study shows that the impaired gasping displayed by neonatal TPH2-/- mice, deficient in CNS serotonin, is restored by 5-HT2A receptor activation. Following 5-HT2A blockade, wild-type mice hypoventilated and their gasping resembled that of TPH2-/- mice. This study shows that both eupnea and gasping in vivo rely on the activation of 5-HT2A receptors.


Subject(s)
Hypoventilation/chemically induced , Receptor, Serotonin, 5-HT2A/physiology , Respiratory Mechanics/physiology , Respiratory Rate/physiology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Animals, Newborn , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Receptor, Serotonin, 5-HT2A/drug effects , Respiratory Mechanics/drug effects , Respiratory Rate/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacology , Tryptophan Hydroxylase
10.
PLoS One ; 16(3): e0248394, 2021.
Article in English | MEDLINE | ID: mdl-33711054

ABSTRACT

We aimed at evaluating the anti-asthmatic effect of cis-[Ru(bpy)2(2-MIM)(NO)](PF6)3 (FOR811A), a nitrosyl-ruthenium compound, in a murine model of allergic asthma. The anti-asthmatic effects were analyzed by measuring the mechanical lung and morphometrical parameters in female Swiss mice allocated in the following groups: untreated control (Ctl+Sal) and control treated with FOR811A (Ctl+FOR), along asthmatic groups untreated (Ast+Sal) and treated with FOR811A (Ast+FOR). The drug-protein interaction was evaluated by in-silico assay using molecular docking. The results showed that the use of FOR811A in experimental asthma (Ast+FOR) decreased the pressure-volume curve, hysteresis, tissue elastance, tissue resistance, and airway resistance, similar to the control groups (Ctl+Sal; Ctl+FOR). However, it differed from the untreated asthmatic group (Ast+Sal, p<0.05), indicating that FOR811A corrected the lung parenchyma and relaxed the smooth muscles of the bronchi. Similar to control groups (Ctl+Sal; Ctl+FOR), FOR811A increased the inspiratory capacity and static compliance in asthmatic animals (Ast+Sal, p<0.05), showing that this metallodrug improved the capacity of inspiration during asthma. The morphometric parameters showed that FOR811A decreased the alveolar collapse and kept the bronchoconstriction during asthma. Beyond that, the molecular docking using FOR811A showed a strong interaction in the distal portion of the heme group of the soluble guanylate cyclase, particularly with cysteine residue (Cys141). In summary, FOR811A relaxed bronchial smooth muscles and improved respiratory mechanics during asthma, providing a protective effect and promising use for the development of an anti-asthmatic drug.


Subject(s)
Anti-Asthmatic Agents , Asthma , Nitric Oxide Donors , Organometallic Compounds , Respiratory Mechanics/drug effects , Ruthenium , Animals , Anti-Asthmatic Agents/chemistry , Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Asthma/physiopathology , Female , Mice , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacology
11.
Respir Res ; 22(1): 71, 2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33637075

ABSTRACT

Delivery of medications to preterm neonates receiving non-invasive ventilation (NIV) represents one of the most challenging scenarios for aerosol medicine. This challenge is highlighted by the undersized anatomy and the complex (patho)physiological characteristics of the lungs in such infants. Key physiological restraints include low lung volumes, low compliance, and irregular respiratory rates, which significantly reduce lung deposition. Such factors are inherent to premature birth and thus can be regarded to as the intrinsic factors that affect lung deposition. However, there are a number of extrinsic factors that also impact lung deposition: such factors include the choice of aerosol generator and its configuration within the ventilation circuit, the drug formulation, the aerosol particle size distribution, the choice of NIV type, and the patient interface between the delivery system and the patient. Together, these extrinsic factors provide an opportunity to optimize the lung deposition of therapeutic aerosols and, ultimately, the efficacy of the therapy.In this review, we first provide a comprehensive characterization of both the intrinsic and extrinsic factors affecting lung deposition in premature infants, followed by a revision of the clinical attempts to deliver therapeutic aerosols to premature neonates during NIV, which are almost exclusively related to the non-invasive delivery of surfactant aerosols. In this review, we provide clues to the interpretation of existing experimental and clinical data on neonatal aerosol delivery and we also describe a frame of measurable variables and available tools, including in vitro and in vivo models, that should be considered when developing a drug for inhalation in this important but under-served patient population.


Subject(s)
Bronchodilator Agents/administration & dosage , Drug Delivery Systems/methods , Nebulizers and Vaporizers , Noninvasive Ventilation/methods , Premature Birth/drug therapy , Respiratory Mechanics/drug effects , Administration, Inhalation , Aerosols , Drug Delivery Systems/instrumentation , Humans , Infant, Newborn , Noninvasive Ventilation/instrumentation , Premature Birth/diagnosis , Premature Birth/physiopathology , Respiratory Mechanics/physiology
12.
Exp Biol Med (Maywood) ; 246(9): 1094-1103, 2021 05.
Article in English | MEDLINE | ID: mdl-33601911

ABSTRACT

Assessment of respiratory mechanics extends from basic research and animal modeling to clinical applications in humans. However, to employ the applications in human models, it is desirable and sometimes mandatory to study non-human animals first. To acquire further precise and controlled signals and parameters, the animals studied must be further distant from their spontaneous ventilation. The majority of respiratory mechanics studies use positive pressure ventilation to model the respiratory system. In this scenario, a few drug categories become relevant: anesthetics, muscle blockers, bronchoconstrictors, and bronchodilators. Hence, the main objective of this study is to briefly review and discuss each drug category, and the impact of a drug on the assessment of respiratory mechanics. Before and during the positive pressure ventilation, the experimental animal must be appropriately sedated and anesthetized. The sedation will lower the pain and distress of the studied animal and the plane of anesthesia will prevent the pain. With those drugs, a more controlled procedure is carried out; further, because many anesthetics depress the respiratory system activity, a minimum interference of the animal's respiration efforts are achieved. The latter phenomenon is related to muscle blockers, which aim to minimize respiratory artifacts that may interfere with forced oscillation techniques. Generally, the respiratory mechanics are studied under appropriate anesthesia and muscle blockage. The application of bronchoconstrictors is prevalent in respiratory mechanics studies. To verify the differences among studied groups, it is often necessary to challenge the respiratory system, for example, by pharmacologically inducing bronchoconstriction. However, the selected bronchoconstrictor, doses, and administration can affect the evaluation of respiratory mechanics. Although not prevalent, studies have applied bronchodilators to return (airway resistance) to the basal state after bronchoconstriction. The drug categories can influence the mathematical modeling of the respiratory system, systemic conditions, and respiratory mechanics outcomes.


Subject(s)
Models, Animal , Respiratory Mechanics/drug effects , Anesthetics/pharmacology , Animals , Bronchoconstrictor Agents/pharmacology , Bronchodilator Agents/pharmacology , Neuromuscular Blocking Agents/pharmacology
13.
Chest ; 159(1): 116-127, 2021 01.
Article in English | MEDLINE | ID: mdl-32603714

ABSTRACT

BACKGROUND: COPD is associated with nighttime respiratory symptoms, poor sleep quality, and increased risk of nocturnal death. Overnight deterioration of inspiratory capacity (IC) and FEV1 have been documented previously. However, the precise nature of this deterioration and mechanisms by which evening bronchodilation may mitigate this occurrence have not been studied. RESEARCH QUESTION: What is the effect of evening dosing of dual, long-acting bronchodilation on detailed nocturnal respiratory mechanics and inspiratory neural drive (IND)? STUDY DESIGN AND METHODS: A double-blind, randomized, placebo-controlled crossover study assessed the effects of evening long-acting bronchodilation (aclidinium bromide/formoterol fumarate dihydrate: 400/12 µg) or placebo on morning trough IC (12 h after the dose; primary outcome) and serial overnight measurements of spirometry, dynamic respiratory mechanics, and IND (secondary outcomes). Twenty participants with COPD (moderate/severe airway obstruction and lung hyperinflation) underwent serial measurements of IC, spirometry, breathing pattern, esophageal and transdiaphragmatic pressures, and diaphragm electromyography (diaphragmatic electromyography as a percentage of maximum; IND) at 6 time points from 0 to 12 h after the dose and compared with sleeping IND. RESULTS: Compared with placebo, evening bronchodilation was not associated with increased morning trough IC 12 h after the dose (P = .48); however, nadir IC (lowest IC, independent of time), peak IC, area under the curve for 12 h after the dose, and IC for 10 h after the dose were improved (P < .05). During placebo, total airways resistance, lung hyperinflation, IND, and tidal esophageal and transdiaphragmatic pressure swings all increased significantly overnight compared with baseline evening values; however, each of these parameters improved with bronchodilator treatment (P < .05) with no change in ventilation or breathing pattern. INTERPRETATION: Respiratory mechanics significantly deteriorated at night during placebo. Although the morning trough IC was unchanged, evening bronchodilator treatment was associated consistently with sustained overnight improvements in dynamic respiratory mechanics and inspiratory neural drive compared with placebo CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov identifier NCT02429765.


Subject(s)
Bronchodilator Agents/administration & dosage , Formoterol Fumarate/administration & dosage , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiratory Mechanics/drug effects , Tropanes/administration & dosage , Aged , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Female , Forced Expiratory Volume/drug effects , Humans , Inspiratory Capacity/drug effects , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/complications , Sleep , Spirometry
14.
J Clin Pharm Ther ; 46(3): 711-716, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33351197

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Propofol provides a prominent sedation effect in gastroscopy. However, sedation with propofol alone during gastroscopy might result in circulatory and respiratory depression. This study aimed to test the hypothesis that the addition of intravenous lidocaine to propofol-based sedation could decrease the ED50 of propofol induction dose during gastroscopy in adult patients while the respiratory and haemodynamic stability were not compromised. METHODS: Patients undergoing gastroscopy were randomly enrolled into lidocaine + propofol (L + P) group or normal saline + propofol (NS + P) group. Subjects were initially administered intravenous bolus of 1.5 mg/kg lidocaine in L + P group or equivalent volume of 0.9% saline in NS + P group. Anaesthesia was then induced with a single bolus of 1.0 µg sufentanil followed by injection of propofol in all patients. The induction dose of propofol for each individual patient was determined by the protocol of Dixon "up-and-down" method for both groups. The primary end point was the ED50 of propofol induction dose. RESULTS: Totally, 48 patients were enrolled and completed this study. Compared with the NS + P group, the ED50 of propofol induction dose was significantly reduced in the L + P group (2.01 mg/kg vs. 1.69 mg/kg) (U = 61.5, p < 0.001). WHAT IS NEW AND CONCLUSION: The addition of intravenous lidocaine significantly reduced the ED50 of propofol induction dose during gastroscopy in adult patients. TRIAL REGISTRATION: The present clinical trial was registered at http://www.chictr.org.cn/ (registration No. ChiCTR1900024025, 23 June 2019).


Subject(s)
Anesthetics, Intravenous/administration & dosage , Gastroscopy/methods , Lidocaine/administration & dosage , Propofol/administration & dosage , Adult , Anesthetics, Intravenous/adverse effects , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Hemodynamics/drug effects , Humans , Male , Middle Aged , Propofol/adverse effects , Prospective Studies , Respiratory Mechanics/drug effects
15.
Intern Emerg Med ; 16(5): 1183-1190, 2021 08.
Article in English | MEDLINE | ID: mdl-33222116

ABSTRACT

The role of noninvasive positive pressure ventilation (NIPPV) in COVID-19 patients with acute hypoxemic respiratory failure (AHRF) is uncertain, as no direct evidence exists to support NIPPV use in such patients. We retrospectively assessed the effectiveness and safety of NIPPV in a cohort of COVID-19 patients consecutively admitted to the COVID-19 general wards of a medium-size Italian hospital, from March 6 to May 7, 2020. Healthcare workers (HCWs) caring for COVID-19 patients were monitored, undergoing nasopharyngeal swab for SARS-CoV-2 in case of onset of COVID-19 symptoms, and periodic SARS-CoV-2 screening serology. Overall, 50 patients (mean age 74.6 years) received NIPPV, of which 22 (44%) were successfully weaned, avoiding endotracheal intubation (ETI) and AHRF-related death. Due to limited life expectancy, 25 (50%) of 50 NIPPV-treated patients received a "do not intubate" (DNI) order. Among these, only 6 (24%) were weaned from NIPPV. Of the remaining 25 NIPPV-treated patients without treatment limitations, 16 (64%) were successfully weaned, 9 (36%) underwent delayed ETI and, of these, 3 (33.3%) died. NIPPV success was predicted by the use of corticosteroids (OR 15.4, CI 1.79-132.57, p 0.013) and the increase in the PaO2/FiO2 ratio measured 24-48 h after NIPPV initiation (OR 1.02, CI 1-1.03, p 0.015), while it was inversely correlated with the presence of a DNI order (OR 0.03, CI 0.001-0.57, p 0.020). During the study period, 2 of 124 (1.6%) HCWs caring for COVID-19 patients were diagnosed with SARS-CoV-2 infection. Apart from patients with limited life expectancy, NIPPV was effective in a substantially high percentage of patients with COVID-19-associated AHRF. The risk of SARS-CoV-2 infection among HCWs was low.


Subject(s)
COVID-19/complications , Noninvasive Ventilation/standards , Positive-Pressure Respiration/standards , Respiratory Insufficiency/etiology , Aged , Aged, 80 and over , COVID-19/therapy , Chi-Square Distribution , Female , Humans , Infection Control/instrumentation , Infection Control/methods , Infection Control/statistics & numerical data , Male , Middle Aged , Multivariate Analysis , Noninvasive Ventilation/methods , Noninvasive Ventilation/statistics & numerical data , Organ Dysfunction Scores , Positive-Pressure Respiration/methods , Positive-Pressure Respiration/statistics & numerical data , Respiratory Insufficiency/therapy , Respiratory Mechanics/drug effects , Respiratory Mechanics/physiology , Retrospective Studies
16.
Sci Rep ; 10(1): 20730, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33244048

ABSTRACT

Pulmonary function testing (PFT) is an important component for evaluating the outcome of experimental rodent models of respiratory diseases. Respiratory inductance plethysmography (RIP) provides a noninvasive method of PFT requiring minimal cooperation. RIP measures work of breathing (WOB) indices including phase angle (Ф), percent rib cage (RC %), breaths per minute (BPM), and labored breathing index (LBI) on an iPad. The aim of this study was to evaluate the utility of a recently developed research instrument, pneuRIP, for evaluation of WOB indices in a developmental rat model. Sprague Dawley rats (2 months old) were commercially acquired and anaesthetised with isoflurane. The pneuRIP system uses two elastic bands: one band (RC) placed around the rib cage under the upper armpit and another band (AB) around the abdomen. The typical thoracoabdominal motion (TAM) plot showed the abdomen and rib cage motion in synchrony. The plots of phase angle and LBI as a function of data point number showed that values were within the range. The distribution for phase angle and LBI was within a narrow range. pneuRIP testing provided instantaneous PFT results. This study demonstrated the utility of RIP as a rapid, noninvasive approach for evaluating treatment interventions in the rodent model.


Subject(s)
Plethysmography/methods , Work of Breathing/physiology , Abdomen/physiology , Animals , Child , Humans , Infant , Isoflurane/administration & dosage , Lung/drug effects , Lung/physiology , Models, Animal , Movement/drug effects , Movement/physiology , Rats , Rats, Sprague-Dawley , Respiration/drug effects , Respiratory Function Tests/methods , Respiratory Mechanics/drug effects , Respiratory Mechanics/physiology , Work of Breathing/drug effects
17.
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
18.
Eur J Pharmacol ; 885: 173514, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32860810

ABSTRACT

PK20M (Dmt-D-Lys-Phe-Phe-OH) is a novel modified endomorphin-2 (EM-2) peptide producing strong dose- and time-dependent antinociceptive activity. Yet its prototype, endogenous EM-2, has been reported to trigger respiratory and vascular effects such as apnea and hypotension. The purpose of this study was to investigate the potency of the PK20M to evoke respiratory and cardiovascular responses in comparison to endogenous endomorphins. The engagement of the vagal pathway and µ opioid receptors in mediation of these responses was investigated. The effects of intravenous injections of PK20M, EM-1, and EM-2 were studied in anaesthetized, spontaneously breathing rats. The main dose-dependent effect of all endomorphins in the intact rats was immediate apnea, blood pressure and heart rate decrease. PK20M produced apnea in at least half of the intact animals in a much smaller dose than EM-1 and EM-2. The effects of all compounds were abrogated by pre-treatment with MNLX, a peripherally acting µ receptor antagonist. Cervical vagotomy eliminated arrest of breathing in the case of each tested compound. Hypotension was reduced by vagi section only after EM-1 and EM-2 administration. Our results demonstrated that apnea and bradycardia caused by systemic injection of all endomorphins were mediated via activation of µ vagal opioid receptors. The hypotension depended on intact vagi nerves only in the case of EM-1 and EM-2, whereas PK20M decreased blood pressure via other mechanisms outside vagal innervation. Modified opioid agonist is more potent in evoking extended hypotension; at the same time, it produces an arrest of breathing less frequently than its prototype EM-2.


Subject(s)
Analgesics/pharmacology , Apnea/chemically induced , Hypotension/chemically induced , Oligopeptides/pharmacology , Opioid Peptides/pharmacology , Vagus Nerve Diseases/chemically induced , Analgesics, Opioid/pharmacology , Animals , Bradycardia/chemically induced , Bradycardia/physiopathology , Dose-Response Relationship, Drug , Injections, Intravenous , Male , Rats , Rats, Wistar , Respiratory Mechanics/drug effects , Vagotomy
19.
J Crit Care ; 60: 50-57, 2020 12.
Article in English | MEDLINE | ID: mdl-32739760

ABSTRACT

Patient ventilatory dyssynchrony (PVD) is a mismatch between the respiratory drive of the patient and ventilatory assistance. It is a complex event seen in almost all ventilated patients and at any ventilator mode, with uncertain significance and prognosis. Due to its different pathophysiological mechanisms, there is still not consensual classification to guide us in selecting the best treatment. In the present review we aimed to summarize some clinical data on PVD, and to propose a clinical classification based on the type of PVD, from potentially innocuous to clearly harmful PVD, which could help clinicians in the decision-making process from adjusting ventilator settings to deeply sedate or paralyze the patient. Clearly, further studies are needed addressing risk factors, physiologic mechanisms and direct consequences of PVD in order to help clinicians to design effective and proven strategies at the bedside.


Subject(s)
Critical Care/methods , Deep Sedation/methods , Hypnotics and Sedatives/administration & dosage , Respiration, Artificial/adverse effects , Respiratory Mechanics/drug effects , Ventilators, Mechanical , Critical Illness , Humans , Maximal Respiratory Pressures , Mental Disorders , Prognosis
20.
Korean J Anesthesiol ; 73(5): 408-416, 2020 10.
Article in English | MEDLINE | ID: mdl-32668833

ABSTRACT

Background: Interscalene brachial plexus block (ISB) provides excellent analgesia for arthroscopic shoulder surgeries but is associated with adverse effects including hemidiaphragmatic paresis. We aimed to compare the respiratory effects, forced vital capacity (FVC), and forced expiratory volume in 1 second (FEV1) between suprascapular nerve block (SSB) and ISB. METHODS: Sixty patients were recruited and randomized into ISB, anterior SSB, and posterior SSB groups. FVC, FEV1, and diaphragmatic excursion were evaluated at baseline and 30 minutes after intervention. Blocks were performed under ultrasound guidance with 15 ml of 0.5% ropivacaine. Pain scores were assessed at 6, 12, and 24 hours postoperatively. RESULTS: The ISB group showed a reduced FVC of 31.2% ± 17.5% (mean ± SD), while the anterior and posterior SSB groups had less reduction of 3.6% ± 18.6% and 6.8% ± 6.5%, respectively (P < 0.001). The ISB group showed more reduction in diaphragmatic excursion than the anterior and posterior SSB groups (median [IQR]): -85.7% (-95.3% to -63.3%) vs. -1.8% (-13.1% to 2.3%) and -1.2% (-8.8% to 16.8%), respectively (P < 0.001). The median pain scores (IQR) in the ISB and anterior SSB groups were lower than those in the posterior SSB group at 6 hours on movement: 0 (0-2), 1.8 (0-4.5) vs. 5 (2.5-8), respectively (P = 0.002). There was no significant difference in oxycodone consumption postoperatively. CONCLUSIONS: Anterior SSB preserves lung function and has a comparable analgesic effect as ISB. Thus, it is recommended for arthroscopic shoulder surgeries, especially in patients who have reduced lung function.


Subject(s)
Analgesia/methods , Arthroscopy/methods , Brachial Plexus Block/methods , Pain, Postoperative/prevention & control , Respiratory Mechanics/physiology , Shoulder/surgery , Adult , Analgesia/adverse effects , Anesthesia, General/adverse effects , Anesthesia, General/methods , Arthroscopy/adverse effects , Brachial Plexus Block/adverse effects , Female , Humans , Male , Middle Aged , Pain, Postoperative/etiology , Respiratory Mechanics/drug effects , Scapula/surgery , Single-Blind Method , Vital Capacity/drug effects , Vital Capacity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...