Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 14.047
Filtrar
1.
BMC Res Notes ; 17(1): 170, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38902794

RESUMO

OBJECTIVE AND RESULTS DESCRIPTION: The study objective was to investigate the potential of quantitative measures of pulmonary inflammation by [18 F]Fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) as a surrogate marker of inflammation in COPD. Patients treated with anti-inflammatory Liraglutide were compared to placebo and correlated with inflammatory markers. 27 COPD-patients (14 receiving Liraglutide treatment and 13 receiving placebo) underwent 4D-respiratory-gated FDG-PET/CT before and after treatment. Two raters independently segmented the lungs from CT images and measured activity in whole lung, mean standard uptake values (SUVmean) corrected for lean-body-mass in the phase-matched PET images of the whole segmented lung volume, and total lesion glycolysis (TLG; SUVmean multiplied by volume). Inter-rater reliability was analyzed with Bland-Altman analysis and correlation plots. We found no differences in metabolic activity in the lungs between the two groups as a surrogate of pulmonary inflammation, and no changes in inflammation markers. The purpose of the research and brief summary of main findings. The degree of and changes in pulmonary inflammation in chronic obstructive pulmonary disease (COPD) may be difficult to ascertain. Measuring metabolic activity as a surrogate marker of inflammation by FDG-PET/CT may be useful, but data on its use in COPD including reproducibility is still limited, especially with respiration-gated technique, which should improve quantification in the lungs. We assessed several quantitative measures of metabolic activity and correlated them with inflammation markers, and we assessed reproducibility of the methods. We found no differences in metabolic activity between the two groups (before and after 40 weeks treatment with Liraglutide vs. placebo). Bland-Altman analysis showed good agreement between the two raters. TRIAL REGISTRATION: The study was conducted between February 2018 and March 2020 at the Department of Pulmonary Diseases at Hospital South West Jutland and Lillebaelt Hospital, Denmark, and registered from March 2018 at clinicaltrials.gov with trial registration number NCT03466021.


Assuntos
Fluordesoxiglucose F18 , Pulmão , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Doença Pulmonar Obstrutiva Crônica , Humanos , Doença Pulmonar Obstrutiva Crônica/diagnóstico por imagem , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Masculino , Feminino , Idoso , Pessoa de Meia-Idade , Pulmão/diagnóstico por imagem , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/metabolismo , Pneumonia/diagnóstico por imagem , Pneumonia/metabolismo , Pneumonia/tratamento farmacológico , Liraglutida/uso terapêutico , Liraglutida/farmacologia , Respiração/efeitos dos fármacos , Compostos Radiofarmacêuticos
2.
Dev Psychobiol ; 66(6): e22518, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38924086

RESUMO

All terrestrial vertebrate life must transition from aquatic gas exchange in the embryonic environment to aerial or pulmonary respiration at birth. In addition to being able to breathe air, neonates must possess functional sensory feedback systems for maintaining acid-base balance. Respiratory neurons in the brainstem act as pH sensors that can adjust breathing to regulate systemic pH. The central pH sensitivity of breathing-related motor output develops over the embryonic period in the zebra finch (Taeniopygia guttata). Due to the key role of chloride ions in electrochemical stability and developmental plasticity, we tested chloride's role in the development of central pH sensitivity. We blocked gamma-aminobutyric acid-A receptors and cation-chloride cotransport that subtly modulated the low-pH effects on early breathing biorhythms. Further, chloride-free artificial cerebrospinal fluid altered the pattern and timing of breathing biorhythms and blocked the stimulating effect of acidosis in E12-14 brainstems. Early and middle stage embryos exhibited rebound plasticity in brainstem motor outputs during low-pH treatment, which was eliminated by chloride-free solution. Results show that chloride modulates low-pH sensitivity and rebound plasticity in the zebra finch embryonic brainstem, but work is needed to determine the cellular and circuit mechanisms that control functional chloride balance during acid-base disturbances.


Assuntos
Tronco Encefálico , Cloretos , Tentilhões , Plasticidade Neuronal , Respiração , Animais , Concentração de Íons de Hidrogênio , Tentilhões/fisiologia , Cloretos/metabolismo , Cloretos/farmacologia , Tronco Encefálico/fisiologia , Tronco Encefálico/efeitos dos fármacos , Respiração/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Embrião não Mamífero/fisiologia
3.
Life Sci ; 351: 122853, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38889841

RESUMO

AIMS: Activation of central respiratory chemoreceptors provides excitatory drive to both respiratory and sympathetic outputs. The enhanced respiratory-sympathetic coupling contributes to the onset and development of hypertension. However, the specific central targets and molecular mechanisms involved in this process remain elusive. This study aimed to investigate the role of acid-sensing ion channel 1 (ASIC1) in nucleus tractus solitarii (NTS) neurons in CO2-stimulated cardiorespiratory effects in spontaneously hypertensive rats (SHRs). MAIN METHODS: Respiration and blood pressure of conscious rats were recorded by whole-body plethysmography and telemetry, respectively. Western blot was used to detect the expression difference of ASIC1 protein in NTS region between Wistar-Kyoto (WKY) rats and SHRs. Excitability of NTS neurons were assessed by extracellular recordings. KEY FINDINGS: Compared to WKY rats, the enhanced CO2-stimulated cardiopulmonary effect and up-regulation of ASIC1 in the NTS were already observed in 4-week-old prehypertensive SHRs. Furthermore, specific blockade of ASIC1 effectively attenuated the CO2-stimulated increase in firing rate of NTS neurons in anesthetized adult SHRs. Intracerebroventricular injections of the ASIC1a blocker PcTx1 or knockdown Asic1 in NTS neurons significantly reduced the heightened CO2-stimulated ventilatory response, and diminished the CO2-stimulated increase in arterial pressure and heart rate in adult SHRs. SIGNIFICANCE: These findings showed that dysregulated ASIC1 signaling in the NTS contribute to the exaggerated CO2-stimulated cardiorespiratory effects observed in SHRs.


Assuntos
Canais Iônicos Sensíveis a Ácido , Pressão Sanguínea , Dióxido de Carbono , Hipertensão , Neurônios , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Núcleo Solitário , Animais , Canais Iônicos Sensíveis a Ácido/metabolismo , Núcleo Solitário/metabolismo , Ratos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Masculino , Dióxido de Carbono/metabolismo , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Pressão Sanguínea/efeitos dos fármacos , Respiração/efeitos dos fármacos , Peptídeos , Venenos de Aranha
4.
Brain Behav Immun ; 119: 333-350, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38561095

RESUMO

Neonatal sepsis remains one of the leading causes of mortality in newborns. Several brainstem-regulated physiological processes undergo disruption during neonatal sepsis. Mechanistic knowledge gaps exist at the interplay between metabolism and immune activation to brainstem neural circuits and pertinent physiological functions in neonates. To delineate this association, we induced systemic inflammation either by TLR4 (LPS) or TLR1/2 (PAM3CSK4) ligand administration in postnatal day 5 mice (PD5). Our findings show that LPS and PAM3CSK4 evoke substantial changes in respiration and metabolism. Physiological trade-offs led to hypometabolic-hypothermic responses due to LPS, but not PAM3CSK4, whereas to both TLR ligands blunted respiratory chemoreflexes. Neuroinflammatory pathways modulation in brainstem showed more robust effects in LPS than PAM3CSK4. Brainstem neurons, microglia, and astrocyte gene expression analyses showed unique responses to TLR ligands. PAM3CSK4 did not significantly modulate gene expression changes in GLAST-1 positive brainstem astrocytes. PD5 pups receiving PAM3CSK4 failed to maintain a prolonged metabolic state repression, which correlated to enhanced gasping latency and impaired autoresuscitation during anoxic chemoreflex challenges. In contrast, LPS administered pups showed no significant changes in anoxic chemoreflex. Electrophysiological studies from brainstem slices prepared from pups exposed to either TLR4 or PAM3CSK4 showed compromised transmission between preBötzinger complex and Hypoglossal as an exclusive response to the TLR1/2 ligand. Spatial gene expression analysis demonstrated a region-specific modulation of PAM3CSK4 within the raphe nucleus relative to other anatomical sites evaluated. Our findings suggest that metabolic changes due to inflammation might be a crucial tolerance mechanism for neonatal sepsis preserving neural control of breathing.


Assuntos
Animais Recém-Nascidos , Tronco Encefálico , Lipopolissacarídeos , Sepse Neonatal , Receptor 1 Toll-Like , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Animais , Camundongos , Receptor 4 Toll-Like/metabolismo , Lipopolissacarídeos/farmacologia , Receptor 2 Toll-Like/metabolismo , Sepse Neonatal/metabolismo , Tronco Encefálico/metabolismo , Receptor 1 Toll-Like/metabolismo , Lipopeptídeos/farmacologia , Respiração/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Astrócitos/metabolismo , Masculino , Ligantes , Microglia/metabolismo , Feminino , Inflamação/metabolismo
5.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38673988

RESUMO

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.


Assuntos
Hipercapnia , Doença de Parkinson , Receptor 5-HT1A de Serotonina , Receptor 5-HT2A de Serotonina , Animais , Masculino , Ratos , Modelos Animais de Doenças , Dopamina/metabolismo , Hipercapnia/metabolismo , Hipercapnia/fisiopatologia , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Oxidopamina/farmacologia , Doença de Parkinson/metabolismo , Ratos Wistar , Receptor 5-HT1A de Serotonina/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Respiração/efeitos dos fármacos , Serotonina/metabolismo , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Agonistas do Receptor 5-HT2 de Serotonina/farmacologia
6.
JAMA ; 328(14): 1405-1414, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36219407

RESUMO

Importance: Opioids can cause severe respiratory depression by suppressing feedback mechanisms that increase ventilation in response to hypercapnia. Following the addition of boxed warnings to benzodiazepine and opioid products about increased respiratory depression risk with simultaneous use, the US Food and Drug Administration evaluated whether other drugs that might be used in place of benzodiazepines may cause similar effects. Objective: To study whether combining paroxetine or quetiapine with oxycodone, compared with oxycodone alone, decreases the ventilatory response to hypercapnia. Design, Setting, and Participants: Randomized, double-blind, crossover clinical trial at a clinical pharmacology unit (West Bend, Wisconsin) with 25 healthy participants from January 2021 through May 25, 2021. Interventions: Oxycodone 10 mg on days 1 and 5 and the following in a randomized order for 5 days: paroxetine 40 mg daily, quetiapine twice daily (increasing daily doses from 100 mg to 400 mg), or placebo. Main Outcomes and Measures: Ventilation at end-tidal carbon dioxide of 55 mm Hg (hypercapnic ventilation) using rebreathing methodology assessed for paroxetine or quetiapine with oxycodone, compared with placebo and oxycodone, on days 1 and 5 (primary) and for paroxetine or quetiapine alone compared with placebo on day 4 (secondary). Results: Among 25 participants (median age, 35 years [IQR, 30-40 years]; 11 female [44%]), 19 (76%) completed the trial. The mean hypercapnic ventilation was significantly decreased with paroxetine plus oxycodone vs placebo plus oxycodone on day 1 (29.2 vs 34.1 L/min; mean difference [MD], -4.9 L/min [1-sided 97.5% CI, -∞ to -0.6]; P = .01) and day 5 (25.1 vs 35.3 L/min; MD, -10.2 L/min [1-sided 97.5% CI, -∞ to -6.3]; P < .001) but was not significantly decreased with quetiapine plus oxycodone vs placebo plus oxycodone on day 1 (33.0 vs 34.1 L/min; MD, -1.2 L/min [1-sided 97.5% CI, -∞ to 2.8]; P = .28) or on day 5 (34.7 vs 35.3 L/min; MD, -0.6 L/min [1-sided 97.5% CI, -∞ to 3.2]; P = .37). As a secondary outcome, mean hypercapnic ventilation was significantly decreased on day 4 with paroxetine alone vs placebo (32.4 vs 41.7 L/min; MD, -9.3 L/min [1-sided 97.5% CI, -∞ to -3.9]; P < .001), but not with quetiapine alone vs placebo (42.8 vs 41.7 L/min; MD, 1.1 L/min [1-sided 97.5% CI, -∞ to 6.4]; P = .67). No drug-related serious adverse events were reported. Conclusions and Relevance: In this preliminary study involving healthy participants, paroxetine combined with oxycodone, compared with oxycodone alone, significantly decreased the ventilatory response to hypercapnia on days 1 and 5, whereas quetiapine combined with oxycodone did not cause such an effect. Additional investigation is needed to characterize the effects after longer-term treatment and to determine the clinical relevance of these findings. Trial Registration: ClinicalTrials.gov Identifier: NCT04310579.


Assuntos
Analgésicos Opioides , Antidepressivos , Oxicodona , Paroxetina , Fumarato de Quetiapina , Insuficiência Respiratória , Adulto , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Antidepressivos/efeitos adversos , Antidepressivos/farmacologia , Benzodiazepinas/efeitos adversos , Benzodiazepinas/farmacologia , Dióxido de Carbono/análise , Método Duplo-Cego , Feminino , Humanos , Hipercapnia/etiologia , Oxicodona/efeitos adversos , Oxicodona/farmacologia , Paroxetina/efeitos adversos , Paroxetina/farmacologia , Fumarato de Quetiapina/efeitos adversos , Fumarato de Quetiapina/farmacologia , Respiração/efeitos dos fármacos , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/diagnóstico
7.
PLoS One ; 17(2): e0263744, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35134094

RESUMO

BACKGROUND: Studies assessing normative values and sex differences in pulmonary function test parameters (PFTPs) among Indigenous populations are sparse. METHODS: PFTPs were compared between male and female Indigenous Australian adults with and without chest radiologically proven chronic airway diseases (CADs). RESULTS: 485 adults (56% were female) with no significant difference in age, body mass index or smoking status between sexes were included. Females displayed a higher prevalence of radiology without CADs compared to males (66 vs. 52%, respectively). Among patients without CADs, after adjustment for age, stature and smoking, males displayed significantly higher absolute values of Forced Vital Capacity (FVC) (mean difference, 0.41L (0.21,0.62), p<0.001) and Forced Expiratory Volume in one second (FEV1) (mean difference 0.27L (0.07,0.47), p<0.001), with no significant difference in FEV1/FVC ratio (mean difference -0.02 (-0.06, 0.02), p = 0.174). Male and female patients with radiologically proven CADs demonstrated lower FEV1/FVC values. However, compared to females, males showed significantly greater reductions in pre- [-0.53 (-0.74, -0.32) vs. -0.29 (-0.42, -0.16), p = 0.045] and post- [-0.51 (-0.72, -0.3) vs. -0.27 (-0.39, -0.14), p = 0.049] bronchodilator FEV1. CONCLUSIONS: There are significant sex differences in the PFTPs among Indigenous Australians. Recognising these differences may be of value in the accurate diagnosis, management, monitoring and prognostication of CADs in this population.


Assuntos
Mecânica Respiratória/fisiologia , Fatores Sexuais , Asma , Austrália/epidemiologia , Broncodilatadores/farmacologia , Feminino , Volume Expiratório Forçado/fisiologia , Humanos , Povos Indígenas , Pulmão/fisiologia , Masculino , Pessoa de Meia-Idade , Prevalência , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Respiração/efeitos dos fármacos , Testes de Função Respiratória/métodos , Caracteres Sexuais , Fumar , Espirometria/métodos , Capacidade Vital/fisiologia
8.
BMC Anesthesiol ; 22(1): 47, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35172747

RESUMO

BACKGROUND: Endoscopic retrograde cholangiopancreatography (ERCP) has been increasingly used to treat patients with biliary/pancreatic duct obstruction or stricture outside the operating room. Effective and safe sedation techniques are needed because of painful stimuli and the long duration of the ERCP procedure.Nalbuphine has been shown to cause less respiratory depression during sedation than similar cases without nalbuphine. This study compared the effects of propofol-nalbuphine (PN) and propofol-fentanyl (PF) sedation in patients undergoing ERCP. METHODS: Four hundred patients scheduled for ERCP procedures were divided into two groups: the PF group (receiving PF sedation,n = 199) and the PN group (receiving PN sedation,n = 201). Vital signs, adverse events during surgery, patient movement scores, pain scores, and adverse events one day post-ERCP were recorded. RESULTS: Stable haemodynamics were observed in both groups.Compared to the PF group, the PN group showed significantly decreased respiratory depression (P < 0.0001) and surgical interruptions (P = 0.048).Nalbuphine decreased patient movement by reducing pain from ERCP. CONCLUSION: Nalbuphine, instead of fentanyl, precipitated less respiratory depression while permitting adequate/equivalent sedation for ERCP and therefore provides more efficient and safer sedation. Trial registration ChiCTR, ChiCTR1800016018, Registered 7 May 2018, http://www.chictr.org.cn/showproj.aspx?proj=27085.


Assuntos
Analgésicos Opioides/farmacologia , Anestésicos Intravenosos/farmacologia , Colangiopancreatografia Retrógrada Endoscópica/métodos , Fentanila/farmacologia , Nalbufina/farmacologia , Propofol/farmacologia , Respiração/efeitos dos fármacos , Adulto , Idoso , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Adulto Jovem
9.
Bull Exp Biol Med ; 172(3): 364-367, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35001305

RESUMO

The article presents a theoretical rationale and a clinical case of relief of post-COVID ventilation failure by inhalation of Xe and O2 gas mixture. Pneumonitis of coronavirus etiology transforms saturated phospholipids of surfactant into a solid-ordered phase, which disrupts surface tension, alveolar pneumatization, and alveolar-capillary gas exchange. Using molecular modeling (B3LYP/lanl2dz; GAUSSIAN09), we demonstrated that Xe atom due to the van der Waals dispersion interaction increases the distance between the phospholipid acyl chains providing a phase transition from the solid-ordered to liquid phase and restored the surface-active monolayer surfactant film. A clinical case confirmed that short-term inhalations of the Xe and O2 gas mixture relieved manifestations of ventilation insufficiency and increased SpO2 and pneumatization of the terminal parts of the lungs.


Assuntos
COVID-19/complicações , Oxigênio/administração & dosagem , Insuficiência Respiratória/terapia , Terapia Respiratória/métodos , Xenônio/administração & dosagem , Administração por Inalação , Anestésicos Inalatórios/administração & dosagem , COVID-19/etiologia , COVID-19/reabilitação , COVID-19/terapia , Combinação de Medicamentos , Humanos , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Respiração/efeitos dos fármacos , Insuficiência Respiratória/etiologia , Federação Russa , SARS-CoV-2 , Síndrome de COVID-19 Pós-Aguda
10.
PLoS One ; 17(1): e0262263, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35030204

RESUMO

PURPOSE: We examined the cardiorespiratory effect of dexmedetomidine, an α2- adrenoceptor/imidazoline 1 (I1) receptor agonist, in spontaneously breathing adult rats. METHODS: Male rats (226-301 g, n = 49) under isoflurane anesthesia had their tail vein cannulated for drug administration and their tail artery cannulated for analysis of mean arterial pressure (MAP), pulse rate (PR), and arterial blood gases (PaO2, PaCO2, pH). After recovery, one set of rats received normal saline for control recording and was then divided into three experimental groups, two receiving dexmedetomidine (5 or 50 µg·kg-1) and one receiving normal saline (n = 7 per group). Another set of rats was divided into four groups receiving dexmedetomidine (50 µg·kg-1) followed 5 min later by 0.5 or 1 mg∙kg-1 atipamezole (selective α2-adrenoceptor antagonist) or efaroxan (α2-adrenoceptor/I1 receptor antagonist) (n = 6 or 8 per group). Recordings were performed 15 min after normal saline or dexmedetomidine administration. RESULTS: Compared with normal saline, dexmedetomidine (5 and 50 µg·kg-1) decreased respiratory frequency (fR, p = 0.04 and < 0.01, respectively), PR (both p < 0.01), and PaO2 (p = 0.04 and < 0.01), and increased tidal volume (both p = 0.049). Dexmedetomidine at 5 µg·kg-1 did not significantly change minute ventilation (V'E) (p = 0.87) or MAP (p = 0.24), whereas dexmedetomidine at 50 µg·kg-1 significantly decreased V'E (p = 0.03) and increased MAP (p < 0.01). Only dexmedetomidine at 50 µg·kg-1 increased PaCO2 (p < 0.01). Dexmedetomidine (5 and 50 µg·kg-1) significantly increased blood glucose (p < 0.01), and dexmedetomidine at 50 µg·kg-1 increased hemoglobin (p = 0.04). Supplemental atipamezole or efaroxan administration similarly prevented the 50 µg·kg-1 dexmedetomidine-related cardiorespiratory changes. PRINCIPAL CONCLUSION: These results suggest that dexmedetomidine-related hypoventilation and hypertension are observed simultaneously and occur predominantly through activation of α2-adrenoceptors, but not I1 receptors, in spontaneously breathing adult rats.


Assuntos
Aptidão Cardiorrespiratória/fisiologia , Dexmedetomidina/farmacologia , Respiração/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Pressão Arterial/efeitos dos fármacos , Benzofuranos/farmacologia , Gasometria/métodos , Pressão Sanguínea/efeitos dos fármacos , Dexmedetomidina/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Hipertensão , Imidazóis/farmacologia , Isoflurano/farmacologia , Masculino , Ratos , Ratos Wistar , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/metabolismo
11.
Am J Perinatol ; 39(2): 204-215, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-32781477

RESUMO

OBJECTIVE: This study aimed to detect which of the two main medicines suggested in the treatment of postligation cardiac syndrome (PLCS)-dobutamine or mirinone-possesses a more therapeutic effect. While doing this, clinicians are provided with a broader perspective on the treatment and follow-up of cases. The desire was to increase the treatability and monitor ability of the cases in question and hence their survivability. STUDY DESIGN: A retrospective review of a cohort of infants with PLCS was conducted between March 2012 and December 2018. In the treatment of infants with PLCS, dobutamine (dobutamine study group-DSG) or milrinone (milrinone study group-MSG) was used. The respiration, cardiac, echocardiography, and perfusion parameters of the cases were assessed both before and after ligation. Based on the data obtained, both the effects of the medicines on PLCS and the difference between their therapeutic effects were studied. The accuracy of prognostication was assessed with receiver operating characteristic analyses. RESULTS: PLCS was detected in 29 (34.1%) of 85 patent ductus arteriosus ligation cases in total. Of all the PLCS cases, 13 (44.8%) were treated with dobutamine and 16 (55.2%) with milrinone. It was observed that the effects of the medicines on the respiratory system and cardiovascular system manifested in the third and 6th hour, respectively. It was detected that both medicines had more effect on the systolic blood pressure (SBP) (area under the curve [AUC]: 0.997/0.996, p = 0.001/0.002) than on the diastolic blood pressure (AUC: 0.911/0.843, p = 0.032/0.046). CONCLUSION: Dobutamine and milrinone, two primary medicines that can be used in the treatment of cases with PLCS, possess similar therapeutic effects on this pathology. In addition, their postoperative therapeutic effects on the SBP are more in the foreground.


Assuntos
Cardiotônicos/administração & dosagem , Sistema Cardiovascular/efeitos dos fármacos , Dobutamina/administração & dosagem , Milrinona/administração & dosagem , Complicações Pós-Operatórias/tratamento farmacológico , Débito Cardíaco/efeitos dos fármacos , Permeabilidade do Canal Arterial/cirurgia , Ecocardiografia , Feminino , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Ligadura , Masculino , Respiração/efeitos dos fármacos , Estudos Retrospectivos , Resultado do Tratamento
12.
J Ethnopharmacol ; 285: 114838, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34788645

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Keguan-1, a new traditional Chinese medicine (TCM) prescription contained seven Chinese herbs, is developed to treat coronavirus disease 19 (COVID-19). The first internationally registered COVID-19 randomised clinical trial on integrated therapy demonstrated that Keguan-1 significantly reduced the incidence of ARDS and inhibited the severe progression of COVID-19. AIM OF THE STUDY: To investigate the protective mechanism of Keguan-1 on ARDS, a lipopolysaccharide (LPS)-induced acute lung injury (ALI) model was used to simulate the pathological state of ARDS in patients with COVID-19, focusing on its effect and mechanism on ALI. MATERIALS AND METHODS: Mice were challenged with LPS (2 mg/kg) by intratracheal instillation (i.t.) and were orally administered Keguan-1 (low dose, 1.25 g/kg; medium dose, 2.5 g/kg; high dose, 5 g/kg) after 2 h. Bronchoalveolar lavage fluid (BALF) and lung tissue were collected 6 h and 24 h after i.t. administration of LPS. The levels of inflammatory factors tumour necrosis factor alpha (TNF-α), interleukin (IL)-6, IL-1ß, keratinocyte-derived chemokine (KC or mCXCL1), macrophage inflammatory protein 2 (MIP2 or mCXCL2), angiotensin II (Ang II), and endothelial cell junction-associated proteins were analysed using ELISA or western blotting. RESULTS: Keguan-1 improved the survival rate, respiratory condition, and pathological lung injury; decreased the production of proinflammatory factors (TNF-α, IL-6, IL-1ß, KC, and MIP2) in BALF and the number of neutrophils in the lung tissues; and ameliorated inflammatory injury in the lung tissues of the mice with LPS-induced ALI. Keguan-1 also reduced the expression of Ang II and the adhesion molecule ICAM-1; increased tight junction proteins (JAM-1 and claudin-5) and VE-cadherin expression; and alleviated pulmonary vascular endothelial injury in LPS-induced ALI. CONCLUSION: These results demonstrate that Keguan-1 can improve LPS-induced ALI by reducing inflammation and pulmonary vascular endothelial injury, providing scientific support for the clinical treatment of patients with COVID-19. Moreover, it also provides a theoretical basis and technical support for the scientific use of TCMs in emerging infectious diseases.


Assuntos
Lesão Pulmonar Aguda , Antivirais/farmacologia , Líquido da Lavagem Broncoalveolar , COVID-19 , Medicamentos de Ervas Chinesas/farmacologia , Pulmão , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/virologia , COVID-19/complicações , COVID-19/imunologia , COVID-19/virologia , Cápsulas , Quimiocina CXCL2/análise , Coix , Forsythia , Interleucina-1beta/análise , Interleucina-6/análise , Lonicera , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Mortalidade , Morus , Fragmentos de Peptídeos/análise , Prunus armeniaca , Respiração/efeitos dos fármacos , SARS-CoV-2 , Resultado do Tratamento , Fator de Necrose Tumoral alfa/análise
13.
Nat Commun ; 12(1): 5962, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34645823

RESUMO

Pitt-Hopkins syndrome (PTHS) is a rare autism spectrum-like disorder characterized by intellectual disability, developmental delays, and breathing problems involving episodes of hyperventilation followed by apnea. PTHS is caused by functional haploinsufficiency of the gene encoding transcription factor 4 (Tcf4). Despite the severity of this disease, mechanisms contributing to PTHS behavioral abnormalities are not well understood. Here, we show that a Tcf4 truncation (Tcf4tr/+) mouse model of PTHS exhibits breathing problems similar to PTHS patients. This behavioral deficit is associated with selective loss of putative expiratory parafacial neurons and compromised function of neurons in the retrotrapezoid nucleus that regulate breathing in response to tissue CO2/H+. We also show that central Nav1.8 channels can be targeted pharmacologically to improve respiratory function at the cellular and behavioral levels in Tcf4tr/+ mice, thus establishing Nav1.8 as a high priority target with therapeutic potential in PTHS.


Assuntos
Haploinsuficiência , Proteínas de Homeodomínio/genética , Hiperventilação/genética , Deficiência Intelectual/genética , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Neurônios/metabolismo , Fator de Transcrição 4/genética , Fatores de Transcrição/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Benzimidazóis/farmacologia , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Dióxido de Carbono/metabolismo , Dióxido de Carbono/farmacologia , Modelos Animais de Doenças , Fácies , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Hiperventilação/tratamento farmacológico , Hiperventilação/metabolismo , Hiperventilação/patologia , Deficiência Intelectual/tratamento farmacológico , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Masculino , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Desempenho Psicomotor/efeitos dos fármacos , Desempenho Psicomotor/fisiologia , Pirazóis/farmacologia , Respiração/efeitos dos fármacos , Fator de Transcrição 4/deficiência , Fatores de Transcrição/metabolismo
14.
Nutrients ; 13(10)2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34684312

RESUMO

The present study aimed to investigate whether acute L-citrulline supplementation would affect inspiratory muscle oxygenation and respiratory performance. Twelve healthy males received 6 g of L-citrulline or placebo in a double-blind crossover design. Pulmonary function (i.e., forced expired volume in 1 s, forced vital capacity and their ratio), maximal inspiratory pressure (MIP), fractional exhaled nitric oxide (NO•), and sternocleidomastoid muscle oxygenation were measured at baseline, one hour post supplementation, and after an incremental resistive breathing protocol to task failure of the respiratory muscles. The resistive breathing task consisted of 30 inspirations at 70% and 80% of MIP followed by continuous inspirations at 90% of MIP until task failure. Sternocleidomastoid muscle oxygenation was assessed using near-infrared spectroscopy. One-hour post-L-citrulline supplementation, exhaled NO• was significantly increased (19.2%; p < 0.05), and this increase was preserved until the end of the resistive breathing (16.4%; p < 0.05). In contrast, no difference was observed in the placebo condition. Pulmonary function and MIP were not affected by the L-citrulline supplementation. During resistive breathing, sternocleidomastoid muscle oxygenation was significantly reduced, with no difference noted between the two supplementation conditions. In conclusion, a single ingestion of 6 g L-citrulline increased NO• bioavailability but not the respiratory performance and inspiratory muscle oxygenation.


Assuntos
Citrulina/farmacologia , Suplementos Nutricionais , Músculos/metabolismo , Óxido Nítrico/metabolismo , Oxigênio/metabolismo , Respiração , Disponibilidade Biológica , Expiração , Volume Expiratório Forçado , Hemoglobinas/metabolismo , Humanos , Masculino , Oxiemoglobinas/metabolismo , Respiração/efeitos dos fármacos , Capacidade Vital
15.
Physiol Rep ; 9(20): e15071, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34699135

RESUMO

Obstructive and central sleep apnea affects ~1 billion people globally and may lead to serious cardiovascular and neurocognitive consequences, but treatment options are limited. High loop gain (ventilatory instability) is a major pathophysiological mechanism underlying both types of sleep apnea and can be lowered pharmacologically with acetazolamide, thereby improving sleep apnea severity. However, individual responses vary and are strongly correlated with the loop gain reduction achieved by acetazolamide. To aid with patient selection for long-term trials and clinical care, our goal was to understand better the factors that determine the change in loop gain following acetazolamide in human subjects with sleep apnea. Thus, we (i) performed several meta-analyses to clarify how acetazolamide affects ventilatory control and loop gain (including its primary components controller/plant gain), and based on these results, we (ii) performed physiological model simulations to assess how different baseline conditions affect the change in loop gain. Our results suggest that (i) acetazolamide primarily causes a left shift of the chemosensitivity line thus lowering plant gain without substantially affecting controller gain; and (ii) higher controller gain, higher paCO2 at eupneic ventilation, and lower CO2 production at baseline result in a more pronounced loop gain reduction with acetazolamide. In summary, the combination of mechanistic meta-analyses with model simulations provides a unified framework of acetazolamide's effects on ventilatory control and revealed physiological predictors of response, which are consistent with empirical observations of acetazolamide's effects in different sleep apnea subgroups. Prospective studies are needed to validate these predictors and assess their value for patient selection.


Assuntos
Acetazolamida/uso terapêutico , Inibidores da Anidrase Carbônica/uso terapêutico , Simulação por Computador , Modelos Biológicos , Respiração/efeitos dos fármacos , Síndromes da Apneia do Sono/tratamento farmacológico , Humanos , Síndromes da Apneia do Sono/patologia
16.
BMC Anesthesiol ; 21(1): 238, 2021 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-34615483

RESUMO

BACKGROUND: Although general anesthetics depress spontaneous respiration, the comprehensive effect of general anesthetics on respiratory function remains unclear. We aimed to investigate the effects of general anesthetics on spontaneous respiration in non-intubated mice with different types and doses of general anesthetic. METHODS: Adult C57BL/6 J mice were administered intravenous anesthetics, including propofol and etomidate, and inhalational anesthetics, including sevoflurane and isoflurane in vivo at doses of 0.5-, 1.0-, and 2.0-times the minimum alveolar concentration (MAC)/median effective dose (ED50) to induce loss of the righting reflex (LORR). Whole-body plethysmography (WBP) was applied to measure parameters of respiration under unrestricted conditions without endotracheal intubation. The alteration in respiratory sensitivity to carbon dioxide (CO2) under general anesthesia was also determined. The following respiratory parameters were continuously recorded during anesthesia or CO2 exposure: respiratory frequency (FR), tidal volume (TV), minute ventilation (MV), expiratory time (TE), inspiratory time (TI), and inspiratory-expiratory time ratio (I/E), and peak inspiratory flow. RESULTS: Sub-anesthetic concentrations (0.5 MAC) of sevoflurane or isoflurane increased FR, TV, and MV. With isoflurane and sevoflurane exposure, the CO2-evoked increases in FR, TV, and MV were decreased. Compared with inhalational anesthetics, propofol and etomidate induced respiratory suppression, affecting FR, TV, and MV. In 100% oxygen (O2), FR in the group that received propofol 1.0-times the ED50 was 69.63 ± 33.44 breaths/min compared with 155.68 ± 64.42 breaths/min in the etomidate-treated group. In the same groups, FR was 88.72 ± 34.51 breaths/min and 225.10 ± 59.82 breaths/min, respectively, in 3% CO2 and 144.17 ± 63.25 breaths/min and 197.70 ± 41.93 breaths/min, respectively, in 5% CO2. A higher CO2 sensitivity was found in etomidate-treated mice compared with propofol-treated mice. In addition, propofol induced a greater decrease in FR, MV, and I/E ratio compared with etomidate, sevoflurane, and isoflurane at equivalent doses (all P < 0.05). CONCLUSIONS: General anesthetics differentially modulate spontaneous breathing in vivo. Volatile anesthetics increase FR, TV, and MV at sub-anesthetic concentrations, while they decrease FR at higher concentrations. Propofol consistently depressed respiratory parameters to a greater degree than etomidate.


Assuntos
Anestésicos Inalatórios/farmacologia , Anestésicos Intravenosos/farmacologia , Propofol/farmacologia , Respiração/efeitos dos fármacos , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Volume de Ventilação Pulmonar/efeitos dos fármacos
17.
Mar Drugs ; 19(9)2021 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-34564149

RESUMO

Fucosylated glycosaminoglycan (FG) from sea cucumber is a potent anticoagulant by inhibiting intrinsic coagulation tenase (iXase). However, high-molecular-weight FGs can activate platelets and plasma contact system, and induce hypotension in rats, which limits its application. Herein, we found that FG from T. ananas (TaFG) and FG from H. fuscopunctata (HfFG) at 4.0 mg/kg (i.v.) could cause significant cardiovascular and respiratory dysfunction in rats, even lethality, while their depolymerized products had no obvious side effects. After injection, native FG increased rat plasma kallikrein activity and levels of the vasoactive peptide bradykinin (BK), consistent with their contact activation activity, which was assumed to be the cause of hypotension in rats. However, the hemodynamic effects of native FG cannot be prevented by the BK receptor antagonist. Further study showed that native FG induced in vivo procoagulation, thrombocytopenia, and pulmonary embolism. Additionally, its lethal effect could be prevented by anticoagulant combined with antiplatelet drugs. In summary, the acute toxicity of native FG is mainly ascribed to pulmonary microvessel embolism due to platelet aggregation and contact activation-mediated coagulation, while depolymerized FG is a safe anticoagulant candidate by selectively targeting iXase.


Assuntos
Anticoagulantes/toxicidade , Glicosaminoglicanos/toxicidade , Animais , Anticoagulantes/química , Coagulação Sanguínea/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Fucose/química , Glicosaminoglicanos/química , Coração/efeitos dos fármacos , Coração/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Ativação Plaquetária/efeitos dos fármacos , Embolia Pulmonar/induzido quimicamente , Embolia Pulmonar/patologia , Ratos Sprague-Dawley , Respiração/efeitos dos fármacos , Pepinos-do-Mar , Função Ventricular Esquerda/efeitos dos fármacos
18.
Am J Physiol Cell Physiol ; 321(4): C681-C683, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34469203

RESUMO

Respiratory depression is a potentially fatal side effect of opioid analgesics and a major limitation to their use. G protein-biased opioid agonists have been proposed as "safer" analgesics with less respiratory depression. These agonists are biased to activate G proteins rather than ß-arrestin signaling. Respiratory depression has been shown to correlate with both G protein bias and intrinsic efficacy, and recent work has refuted the role of ß-arrestin signaling in opioid-induced respiratory depression. In addition, there is substantial evidence that G proteins do, in fact, mediate respiratory depression by actions in respiratory-controlling brainstem neurons. Based on these studies, we provide the perspective that protection from respiratory depression displayed by newly developed G protein-biased agonists is due to factors other than G protein versus arrestin bias.


Assuntos
Analgésicos Opioides/efeitos adversos , Proteínas de Ligação ao GTP/agonistas , Pulmão/efeitos dos fármacos , Respiração/efeitos dos fármacos , Insuficiência Respiratória/induzido quimicamente , beta-Arrestina 2/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/fisiopatologia , Insuficiência Respiratória/metabolismo , Insuficiência Respiratória/fisiopatologia , Insuficiência Respiratória/prevenção & controle , Fatores de Risco , Transdução de Sinais
19.
J Ethnopharmacol ; 280: 114488, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34358653

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Traditional Chinese medicine (TCM) has a long history in the prevention and treatment of pandemics. The TCM formula Lung Cleansing and Detoxifying Decoction (LCDD), also known as Qing Fei Pai Du Decoction, has been demonstrated effective against Coronavirus Disease 2019 (COVID-19). AIM OF THE STUDY: This work aimed to elucidate the active ingredients, targets and pathway mechanism of LCDD related to suppression of inflammatory, immunity regulation and relaxation of airway smooth muscle for the treatment of COVID-19. MATERIALS AND METHODS: Mining chemical ingredients reported in LCDD, 144 compounds covering all herbs were selected and screened against inflammatory-, immunity- and respiratory-related GPCRs including GPR35, H1, CB2, B2, M3 and ß2-adrenoceptor receptor using a label-free integrative pharmacology method. Further, all active compounds were detected using liquid chromatography-tandem mass spectrometry, and an herb-compound-target network based on potency and content of compounds was constructed to elucidate the multi-target and synergistic effect. RESULTS: Thirteen compounds were identified as GPR35 agonists, including licochalcone B, isoliquiritigenin, etc. Licochalcone B, isoliquiritigenin and alisol A exhibited bradykinin receptor B2 antagonism activities. Atractyline and shogaol showed as a cannabinoid receptor CB2 agonist and a histamine receptor H1 antagonist, respectively. Tectorigenin and aristofone acted as muscarinic receptor M3 antagonists, while synephrine, ephedrine and pseudoephedrine were ß2-adrenoceptor agonists. Pathway deconvolution assays suggested activation of GPR35 triggered PI3K, MEK, JNK pathways and EGFR transactivation, and the activation of ß2-adrenoceptor mediated MEK and Ca2+. The herb-compound-target network analysis found that some compounds such as licochalcone B acted on multiple targets, and multiple components interacted with the same target such as GPR35, reflecting the synergistic mechanism of Chinese medicine. At the same time, some low-abundance compounds displayed high target activity, meaning its important role in LCDD for anti-COVID-19. CONCLUSIONS: This study elucidates the active ingredients, targets and pathways of LCDD. This is useful for elucidating multitarget synergistic action for its clinical therapeutic efficacy.


Assuntos
Técnicas Biossensoriais/métodos , Tratamento Farmacológico da COVID-19 , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacologia , Animais , Linhagem Celular Tumoral , Chalconas/farmacologia , Cricetulus , Medicamentos de Ervas Chinesas/análise , Efedrina/farmacologia , Células HEK293 , Humanos , Imunidade/efeitos dos fármacos , Inflamação/metabolismo , Pneumopatias/metabolismo , Músculo Liso/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Respiração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
20.
Nitric Oxide ; 116: 7-13, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34400339

RESUMO

BACKGROUND: Inhaled nitric oxide (NO) is a selective pulmonary vasodilator. In-vitro studies report that NO donors can inhibit replication of SARS-CoV-2. This multicenter study evaluated the feasibility and effects of high-dose inhaled NO in non-intubated spontaneously breathing patients with Coronavirus disease-2019 (COVID-19). METHODS: This is an interventional study to determine whether NO at 160 parts-per-million (ppm) inhaled for 30 min twice daily might be beneficial and safe in non-intubated COVID-19 patients. RESULTS: Twenty-nine COVID-19 patients received a total of 217 intermittent inhaled NO treatments for 30 min at 160 ppm between March and June 2020. Breathing NO acutely decreased the respiratory rate of tachypneic patients and improved oxygenation in hypoxemic patients. The maximum level of nitrogen dioxide delivered was 1.5 ppm. The maximum level of methemoglobin (MetHb) during the treatments was 4.7%. MetHb decreased in all patients 5 min after discontinuing NO administration. No adverse events during treatment, such as hypoxemia, hypotension, or acute kidney injury during hospitalization occurred. In our NO treated patients, one patient of 29 underwent intubation and mechanical ventilation, and none died. The median hospital length of stay was 6 days [interquartile range 4-8]. No discharged patients required hospital readmission nor developed COVID-19 related long-term sequelae within 28 days of follow-up. CONCLUSIONS: In spontaneous breathing patients with COVID-19, the administration of inhaled NO at 160 ppm for 30 min twice daily promptly improved the respiratory rate of tachypneic patients and systemic oxygenation of hypoxemic patients. No adverse events were observed. None of the subjects was readmitted or had long-term COVID-19 sequelae.


Assuntos
Tratamento Farmacológico da COVID-19 , Hospitalização , Óxido Nítrico/administração & dosagem , Pneumonia Viral/tratamento farmacológico , Respiração/efeitos dos fármacos , Administração por Inalação , COVID-19/complicações , COVID-19/virologia , Relação Dose-Resposta a Droga , Humanos , Óxido Nítrico/farmacologia , Óxido Nítrico/uso terapêutico , Pneumonia Viral/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...