Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.807
Filtrar
1.
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
2.
Sci Rep ; 11(1): 24097, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34916560

RESUMO

Levosimendan exerts positive inotropic and vasodilatory effects. Currently, its effects on right heart function remain uncertain. This systematic review and meta-analysis is intended to illustrate the impacts of levosimendan on systolic function of the right heart in patients with heart dysfunction. We systematically searched electronic databases (PubMed, the Cochrane Library, Embase and Web of Science) up to November 30, 2020, and filtered eligible studies that reported the impacts of levosimendan on right heart function. Of these, only studies whose patients suffered from heart dysfunction or pulmonary hypertension were included. Additionally, patients were divided into two groups (given levosimendan or not) in the initial research. Then, RevMan5.3 was used to conduct further analysis. A total of 8 studies comprising 390 patients were included. The results showed that after 24 h of levosimendan, patients' right ventricular fractional area change [3.17, 95% CI (2.03, 4.32), P < 0.00001], tricuspid annular plane systolic excursion [1.26, 95% CI (0.35, 2.16), P = 0.007] and tricuspid annular peak systolic velocity [0.86, 95% CI (0.41, 1.32), P = 0.0002] were significantly increased compared to the control group. And there is an increasing trend of cardiac output in levosimendan group [1.06, 95% CI (- 0.16, 2.29), P = 0.09 ] .Furthermore, patients' systolic pulmonary arterial pressure [- 5.57, 95% CI (- 7.60, - 3.54), P < 0.00001] and mean pulmonary arterial pressure [- 1.01, 95% CI (- 1.64, - 0.37), P = 0.002] were both significantly decreased, whereas changes in pulmonary vascular resistance [- 55.88, 95% CI (- 206.57, 94.82), P = 0.47] were not significant. Our study shows that in patients with heart dysfunction, levosimendan improves systolic function of the right heart and decreases the pressure of the pulmonary artery.


Assuntos
Cardiotônicos , Simendana/administração & dosagem , Simendana/farmacologia , Vasodilatadores , Disfunção Ventricular Direita/tratamento farmacológico , Idoso , Débito Cardíaco/efeitos dos fármacos , Feminino , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/fisiopatologia , Masculino , Pessoa de Meia-Idade , Sístole/efeitos dos fármacos , Resistência Vascular/efeitos dos fármacos , Disfunção Ventricular Direita/fisiopatologia , Função Ventricular Direita/efeitos dos fármacos
3.
Vet Anaesth Analg ; 48(5): 654-662, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34332897

RESUMO

OBJECTIVE: To evaluate the systemic cardiovascular effects of dose escalating administration of norepinephrine in healthy dogs anesthetized with isoflurane. STUDY DESIGN: Experimental study. ANIMALS: A total of six adult laboratory Beagle dogs, 10.5 (9.2-12.0) kg [median (range)]. METHODS: Each dog was anesthetized with isoflurane at an end-tidal concentration of 1.7%, mechanically ventilated and administered a continuous rate infusion of rocuronium (0.5 mg kg-1 hour-1). Each dog was administered incremental dose rates of norepinephrine (0.05, 0.125, 0.25, 0.5, 1.0 and 2.0 µg kg-1 minute-1), and each dose was infused for 15 minutes. Cardiovascular variables were recorded before administration and at the end of each infusion period. RESULTS: Norepinephrine infusion increased mean arterial pressure (MAP), cardiac output (CO) and oxygen delivery in a dose-dependent manner. Systemic vascular resistance did not significantly change during the experiment. Stroke volume increased at the lower dose rates and heart rate increased at the higher dose rates. Oxygen consumption and lactate concentrations did not significantly change during infusions. CONCLUSIONS: In dogs anesthetized with isoflurane, norepinephrine increased MAP by increasing the CO. CO increased with a change in stroke volume at lower dose rates of norepinephrine. At higher dosage, heart rate also contributed to an increase in CO. Norepinephrine did not cause excessive vasoconstriction that interfered with the CO during this study. CLINICAL RELEVANCE: Norepinephrine can be useful for treating hypotension in dogs anesthetized with isoflurane.


Assuntos
Anestésicos Inalatórios , Isoflurano , Norepinefrina , Anestésicos Inalatórios/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Débito Cardíaco/efeitos dos fármacos , Cães , Frequência Cardíaca/efeitos dos fármacos , Isoflurano/farmacologia , Norepinefrina/farmacologia
4.
Environ Toxicol Pharmacol ; 87: 103723, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34391906

RESUMO

Living organisms are commonly exposed to cadmium and other toxic metals. A vast body of research has shown the significant effects of these toxic metals on developmental processes. In order to study the role of toxic metals on early developmental stages of eukaryotes, we explored the effect of cadmium (Cd2+) contaminant on zebrafish. Thus, zebrafish embryos were exposed to 3 mg/L (16.7 µM) Cd2+ for 96 h and imaged every 24 h from the exposure onwards. Hatching rates of the eggs were determined at 72 h, followed by analyses at 96 h for: survival rate, morphometrical factors, and functional parameters of the cardiovascular system. Interestingly enough, significant hatching delays along with smaller cephalic region and some morphological abnormalities were observed in the treatment group. Moreover, substantial changes were noticed in the length of notochord and embryo, absorption of yolk sac with shorter extension, area of swimming bladder, as well as pericardium sac after Cd2+ treatment. Cadmium also caused significant abnormalities in heart physiology which could be the leading cause of mentioned morphological deformities. Herein, our results shine light on systematic acute embryological effects of cadmium in the early development of zebrafish for the first time.


Assuntos
Anormalidades Induzidas por Medicamentos , Anormalidades Múltiplas/induzido quimicamente , Cádmio/toxicidade , Embrião não Mamífero/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Teratógenos/toxicidade , Poluentes Químicos da Água/toxicidade , Anormalidades Induzidas por Medicamentos/fisiopatologia , Animais , Débito Cardíaco/efeitos dos fármacos , Embrião não Mamífero/anormalidades , Embrião não Mamífero/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Volume Sistólico/efeitos dos fármacos , Peixe-Zebra/anormalidades , Peixe-Zebra/fisiologia
5.
Sci Rep ; 11(1): 15849, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34349162

RESUMO

Extracorporeal membrane oxygenation (ECMO) is a life-saving intervention for patients suffering from respiratory or cardiac failure. The ECMO-associated morbidity and mortality depends to a large extent on the underlying disease and is often related to systemic inflammation, consecutive immune paralysis and sepsis. Here we tested the hypothesis that human α1-antitrypsin (SERPINA1) due to its anti-protease and anti-inflammatory functions may attenuate ECMO-induced inflammation. We specifically aimed to test whether intravenous treatment with α1-antitrypsin reduces the release of cytokines in response to 2 h of experimental ECMO. Adult rats were intravenously infused with α1-antitrypsin immediately before starting veno-arterial ECMO. We measured selected pro- and anti-inflammatory cytokines and found, that systemic levels of tumor necrosis factor-α, interleukin-6 and interleukin-10 increase during experimental ECMO. As tachycardia and hypertension developed in response to α1-antitrypsin, a single additional bolus of fentanyl and midazolam was given. Treatment with α1-antitrypsin and higher sedative doses reduced all cytokine levels investigated. We suggest that α1-antitrypsin might have the potential to protect against both ECMO-induced systemic inflammation and immune paralysis. More studies are needed to corroborate our findings, to clarify the mechanisms by which α1-antitrypsin inhibits cytokine release in vivo and to explore the potential application of α1-antitrypsin in clinical ECMO.


Assuntos
Débito Cardíaco/efeitos dos fármacos , Citocinas/metabolismo , Oxigenação por Membrana Extracorpórea/métodos , Hemodinâmica , Inibidores da Tripsina/farmacologia , alfa 1-Antitripsina/farmacologia , Animais , Masculino , Ratos , Ratos Endogâmicos Lew
6.
Crit Care ; 25(1): 302, 2021 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-34419120

RESUMO

BACKGROUND: Through venous contraction, norepinephrine (NE) increases stressed blood volume and mean systemic pressure (Pms) and exerts a "fluid-like" effect. When both fluid and NE are administered, Pms may not only result from the sum of the effects of both drugs. Indeed, norepinephrine may enhance the effects of volume expansion: because fluid dilutes into a more constricted, smaller, venous network, fluid may increase Pms to a larger extent at a higher than at a lower dose of NE. We tested this hypothesis, by mimicking the effects of fluid by passive leg raising (PLR). METHODS: In 30 septic shock patients, norepinephrine was decreased to reach a predefined target of mean arterial pressure (65-70 mmHg by default, 80-85 mmHg in previously hypertensive patients). We measured the PLR-induced increase in Pms (heart-lung interactions method) under high and low doses of norepinephrine. Preload responsiveness was defined by a PLR-induced increase in cardiac index ≥ 10%. RESULTS: Norepinephrine was decreased from 0.32 [0.18-0.62] to 0.26 [0.13-0.50] µg/kg/min (p < 0.0001). This significantly decreased the mean arterial pressure by 10 [7-20]% and Pms by 9 [4-19]%. The increase in Pms (∆Pms) induced by PLR was 13 [9-19]% at the higher dose of norepinephrine and 11 [6-16]% at the lower dose (p < 0.0001). Pms reached during PLR at the high dose of NE was higher than expected by the sum of Pms at baseline at low dose, ∆Pms induced by changing the norepinephrine dose and ∆Pms induced by PLR at low dose of NE (35.6 [11.2] mmHg vs. 33.6 [10.9] mmHg, respectively, p < 0.01). The number of preload responders was 8 (27%) at the high dose of NE and 15 (50%) at the low dose. CONCLUSIONS: Norepinephrine enhances the Pms increase induced by PLR. These results suggest that a bolus of fluid of the same volume has a greater haemodynamic effect at a high dose than at a low dose of norepinephrine during septic shock.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Norepinefrina/farmacologia , Choque Séptico/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Pressão Sanguínea/fisiologia , Débito Cardíaco/efeitos dos fármacos , Feminino , Hemodinâmica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Norepinefrina/administração & dosagem , Norepinefrina/farmacocinética , Substitutos do Plasma/administração & dosagem , Substitutos do Plasma/farmacocinética , Substitutos do Plasma/farmacologia , Choque Séptico/fisiopatologia , Resultado do Tratamento , Vasoconstritores/administração & dosagem , Vasoconstritores/farmacocinética , Vasoconstritores/farmacologia
7.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R385-R395, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34259041

RESUMO

Exercise intolerance is a hallmark symptom of cardiovascular disease and likely occurs via enhanced activation of muscle metaboreflex-induced vasoconstriction of the heart and active skeletal muscle which, thereby limits cardiac output and peripheral blood flow. Muscle metaboreflex vasoconstrictor responses occur via activation of metabolite-sensitive afferent fibers located in ischemic active skeletal muscle, some of which express transient receptor potential vanilloid 1 (TRPV1) cation channels. Local cardiac and intrathecal administration of an ultrapotent noncompetitive, dominant negative agonist resiniferatoxin (RTX) can ablate these TRPV1-sensitive afferents. This technique has been used to attenuate cardiac sympathetic afferents and nociceptive pain. We investigated whether intrathecal administration (L4-L6) of RTX (2 µg/kg) could chronically attenuate subsequent muscle metaboreflex responses elicited by reductions in hindlimb blood flow during mild exercise (3.2 km/h) in chronically instrumented conscious canines. RTX significantly attenuated metaboreflex-induced increases in mean arterial pressure (27 ± 5.0 mmHg vs. 6 ± 8.2 mmHg), cardiac output (1.40 ± 0.2 L/min vs. 0.28 ± 0.1 L/min), and stroke work (2.27 ± 0.2 L·mmHg vs. 1.01 ± 0.2 L·mmHg). Effects were maintained until 78 ± 14 days post-RTX at which point the efficacy of RTX injection was tested by intra-arterial administration of capsaicin (20 µg/kg). A significant reduction in the mean arterial pressure response (+45.7 ± 6.5 mmHg pre-RTX vs. +19.7 ± 3.1 mmHg post-RTX) was observed. We conclude that intrathecal administration of RTX can chronically attenuate the muscle metaboreflex and could potentially alleviate enhanced sympatho-activation observed in cardiovascular disease states.


Assuntos
Débito Cardíaco/efeitos dos fármacos , Diterpenos/farmacologia , Membro Posterior/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Animais , Pressão Arterial/efeitos dos fármacos , Débito Cardíaco/fisiologia , Diterpenos/administração & dosagem , Cães , Coração/efeitos dos fármacos , Coração/fisiopatologia , Membro Posterior/fisiopatologia , Isquemia/fisiopatologia , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiopatologia , Vasoconstrição/fisiologia
8.
Toxicol Appl Pharmacol ; 427: 115652, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34298059

RESUMO

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.


Assuntos
Débito Cardíaco/efeitos dos fármacos , Cardiotônicos/administração & dosagem , Inseticidas/envenenamento , Milrinona/administração & dosagem , Fosfinas/envenenamento , Mecânica Respiratória/efeitos dos fármacos , Animais , Débito Cardíaco/fisiologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Exposição por Inalação/efeitos adversos , Dose Letal Mediana , Profilaxia Pré-Exposição/métodos , Ratos , Ratos Sprague-Dawley , Mecânica Respiratória/fisiologia , Taxa de Sobrevida/tendências
9.
Br J Anaesth ; 127(1): 23-31, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33896589

RESUMO

BACKGROUND: Ketamine has cardiac excitatory side-effects. Currently, data on the effects of ketamine and metabolite concentrations on cardiac output are scarce. We therefore developed a pharmacodynamic model derived from data from a randomised clinical trial. The current study is part of a larger clinical study evaluating the potential mitigating effect of sodium nitroprusside on the psychedelic effects of ketamine. METHODS: Twenty healthy male subjects received escalating esketamine and racemic ketamine doses in combination with either placebo or sodium nitroprusside on four visits: (i) esketamine and placebo, (ii) esketamine and sodium nitroprusside, (iii) racemic ketamine and placebo, and (iv) racemic ketamine and sodium nitroprusside. During each visit, arterial blood samples were obtained and cardiac output was measured. Nonlinear mixed-effect modelling was used to analyse the cardiac output time-series data. Ketamine metabolites were added to the model in a sequential manner to evaluate the effects of metabolites. RESULTS: A model including an S-ketamine and S-norketamine effect best described the data. Ketamine increased cardiac output, whereas modelling revealed that S-norketamine decreased cardiac output. No significant effects were detected for R-ketamine, metabolites other than S-norketamine, or sodium nitroprusside on cardiac output. CONCLUSIONS: S-Ketamine, but not R-ketamine, increased cardiac output in a dose-dependent manner. In contrast to S-ketamine, its metabolite S-norketamine reduced cardiac excitation in a dose-dependent manner. CLINICAL TRIAL REGISTRATION: Dutch Cochrane Center 5359.


Assuntos
Anestésicos Dissociativos/química , Anestésicos Dissociativos/farmacocinética , Débito Cardíaco/efeitos dos fármacos , Ketamina/química , Ketamina/farmacocinética , Adulto , Débito Cardíaco/fisiologia , Estudos Cross-Over , Relação Dose-Resposta a Droga , Método Duplo-Cego , Voluntários Saudáveis , Humanos , Masculino , Estereoisomerismo , Adulto Jovem
10.
Medicine (Baltimore) ; 100(17): e25492, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33907098

RESUMO

ABSTRACT: rbBNP has positive cardiac effects in patients with acute decompensated heart failure, but its effects on the systemic venous circulation are not known.A single-center retrospective, self-controlled study was conducted on 14 patients undergone recombinant human brain natriuretic peptide (rhBNP) treatment between January 1, 2015 to December 31, 2018.The cardiac output (CO) significantly increased from 3.75 ±â€Š1.14 L min-1 to 4.24 ±â€Š0.97 L min-1 30 minutes after rbBNP infusion, and to 4.20 ±â€Š1.19 L min-1 3 hours later. The systemic vascular resistance significantly decreased from 18.85 ±â€Š7.66 mm Hg min L-1 to 14.62 ±â€Š6.13 mm Hg min L-1 30 minutes. The resistance to venous return (VR) significantly decreased from 5.93 ±â€Š4.97 mm Hg min L-1 to 4.46 ±â€Š1.53 mmHg min L-1 3 hours later. The mean systemic filling pressure significantly decreased from 32.71 ±â€Š20.00 mm Hg to 28.254 ±â€Š6.09 mm Hg 3 hours later.The role of rhBNP on CO was to reduce the peripheral circulation resistance at 30 minutes after rhBNP infusion and to reduce the resistance to VR at 3 hours later.This trial is registered at ChiCTR: ID ChiCTR1900024562.


Assuntos
Débito Cardíaco/efeitos dos fármacos , Insuficiência Cardíaca/tratamento farmacológico , Modelos Cardiovasculares , Peptídeo Natriurético Encefálico/uso terapêutico , Resistência Vascular/efeitos dos fármacos , Doença Aguda , Idoso , Feminino , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/uso terapêutico , Estudos Retrospectivos , Resultado do Tratamento
11.
Int J Mol Sci ; 22(6)2021 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-33806919

RESUMO

Argon inhalation attenuates multiorgan failure (MOF) after experimental ischemic injury. We hypothesized that this protection could involve decreased High Mobility Group Box 1 (HMGB1) systemic release. We investigated this issue in an animal model of MOF induced by aortic cross-clamping. Anesthetized rabbits were submitted to supra-coeliac aortic cross-clamping for 30 min, followed by 300 min of reperfusion. They were randomly divided into three groups (n = 7/group). The Control group inhaled nitrogen (70%) and oxygen (30%). The Argon group was exposed to a mixture of argon (70%) and oxygen (30%). The last group inhaled nitrogen/oxygen (70/30%) with an administration of the HMGB1 inhibitor glycyrrhizin (4 mg/kg i.v.) 5 min before aortic unclamping. At the end of follow-up, cardiac output was significantly higher in Argon and Glycyrrhizin vs. Control (60 ± 4 and 49 ± 4 vs. 33 ± 8 mL/kg/min, respectively). Metabolic acidosis was attenuated in Argon and Glycyrrhizin vs. Control, along with reduced amount of norepinephrine to reverse arterial hypotension. This was associated with reduced interleukin-6 and HMGB1 plasma concentration in Argon and Glycyrrhizin vs. Control. End-organ damages were also attenuated in the liver and kidney in Argon and Glycyrrhizin vs. Control, respectively. Argon inhalation reduced HMGB1 blood level after experimental aortic cross-clamping and provided similar benefits to direct HMGB1 inhibition.


Assuntos
Argônio/farmacologia , Proteína HMGB1/antagonistas & inibidores , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Insuficiência de Múltiplos Órgãos/metabolismo , Animais , Biópsia , Pressão Sanguínea/efeitos dos fármacos , Débito Cardíaco/efeitos dos fármacos , Citocinas/sangue , Modelos Animais de Doenças , Testes de Função Cardíaca , Hemodinâmica/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Insuficiência de Múltiplos Órgãos/diagnóstico , Insuficiência de Múltiplos Órgãos/etiologia , Coelhos
12.
Am J Obstet Gynecol ; 225(5): 544.e1-544.e9, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33887239

RESUMO

BACKGROUND: Nifedipine is a widely used drug in pregnancies complicated by maternal hypertensive disorders that can be associated with placental insufficiency and fetal hypoxemia. The evidence regarding fetal myocardial responses to nifedipine in hypoxemia is limited. OBJECTIVE: We hypothesized that nifedipine would not impair fetal sheep cardiac function under hypoxemic environment. In particular, we investigated the effects of nifedipine on fetal ventricular functional parameters and cardiac output. STUDY DESIGN: A total of 21 chronically instrumented fetal sheep at 122 to 134 gestational days (term, 145 days) were included in this study. Fetal cardiac function was evaluated by measuring global longitudinal strain, indices describing ventricular systolic and diastolic function, and cardiac outputs using two-dimensional speckle tracking and tissue and spectral pulsed-wave Doppler echocardiography. Fetal carotid artery blood pressure and blood gas values were invasively monitored. After baseline data collection, fetal hypoxemia was induced by maternal hyperoxygenation. After hypoxemia phase data collection, 9 fetuses received nifedipine infusion, and 12 fetuses received saline infusion. Data were collected 30 and 120 minutes after the infusion was started. After 120 minutes of data collection, maternal and fetal oxygenation were normalized, and normoxemia phase data were collected, while infusion was continued. RESULTS: Hypoxemia decreased fetal carotid artery mean arterial pressure from 40 (8) mm Hg to 35 (8) mm Hg (P<.007), and left ventricular global longitudinal strain showed less deformation than at baseline (P=.001). Under hypoxemia, nifedipine caused a reduction in right ventricular global longitudinal strain (P<.05), a decrease in right ventricular isovolumic relaxation velocity and its deceleration (P<.01) indicating diastolic dysfunction, and a drop in right ventricular cardiac output (P<.05). Nifedipine did not alter fetal left ventricular functional parameters or cardiac output. When normoxemia was restored, fetal right ventricular functional parameters and cardiac output returned to baseline level. CONCLUSION: In hypoxemic fetus, nifedipine impaired right ventricular function and reduced its cardiac output. The detrimental effects of nifedipine on fetal right ventricular function were abolished, when normoxemia was restored. Our findings suggest that in a hypoxemic environment nifedipine triggers detrimental effects on fetal right ventricular function.


Assuntos
Bloqueadores dos Canais de Cálcio/efeitos adversos , Débito Cardíaco/efeitos dos fármacos , Hipóxia Fetal/complicações , Nifedipino/efeitos adversos , Disfunção Ventricular Direita/induzido quimicamente , Animais , Pressão Arterial/efeitos dos fármacos , Artérias Carótidas/efeitos dos fármacos , Diástole/efeitos dos fármacos , Ecocardiografia Doppler de Pulso , Monitorização Fetal , Modelos Animais , Ovinos
13.
Hypertension ; 77(5): 1591-1599, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33775123
14.
Heart Vessels ; 36(7): 1088-1097, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33763729

RESUMO

To characterize in vivo anti-atrial fibrillatory potential and pharmacological safety profile of ranolazine having INa,L plus IKr inhibitory actions in comparison with those of clinically available anti-atrial fibrillatory drugs; namely, dronedarone, amiodarone, bepridil and dl-sotalol in our previous studies, ranolazine dihydrochloride in sub-therapeutic (0.3 mg/kg) and supra-therapeutic (3 mg/kg) doses was intravenously infused over 10 min to the halothane-anesthetized dogs (n = 5). The low dose increased the heart rate, cardiac output and atrioventricular conduction velocity possibly via vasodilator action-induced, reflex-mediated increase of adrenergic tone. Meanwhile, the high dose decreased the heart rate, ventricular contraction, cardiac output and mean blood pressure, indicating that drug-induced direct actions may exceed the reflex-mediated compensation. In addition, it prolonged the atrial and ventricular effective refractory periods, of which potency and selectivity for the former were less great compared with those of the clinically-available drugs. Moreover, it did not alter the ventricular early repolarization period in vivo, but prolonged the late repolarization with minimal risk for re-entrant arrhythmias. These in vivo findings of ranolazine suggest that INa,L suppression may attenuate IKr inhibition-associated prolongation of early repolarization in the presence of reflex-mediated increase of adrenergic tone. Thus, ranolazine alone may be less promising as an anti-atrial fibrillatory drug, but its potential risk for inducing torsade de pointes will be small. These information can be used as a guide to predict the utility and adverse effects of anti-atrial fibrillatory drugs having multi-channel modulatory action.


Assuntos
Anestesia por Inalação/métodos , Fibrilação Atrial/tratamento farmacológico , Halotano/farmacologia , Átrios do Coração/fisiopatologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Ranolazina/administração & dosagem , Potenciais de Ação/efeitos dos fármacos , Anestésicos Inalatórios/farmacologia , Animais , Fibrilação Atrial/fisiopatologia , Débito Cardíaco/efeitos dos fármacos , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Eletrocardiografia/efeitos dos fármacos , Feminino , Átrios do Coração/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiopatologia , Infusões Intravenosas , Bloqueadores dos Canais de Sódio/administração & dosagem
15.
Shock ; 56(3): 352-359, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33756500

RESUMO

ABSTRACT: We performed a systematic review to investigate the effects of vasopressor-induced hemodynamic changes in adults with shock. We applied a physiological approach using the interacting domains of intravascular volume, heart pump performance, and vascular resistance to structure the interpretation of responses to vasopressors. We hypothesized that incorporating changes in determinants of cardiac output and vascular resistance better reflect the vasopressor responsiveness beyond mean arterial pressure alone.We identified 28 studies including 678 subjects in Pubmed, EMBASE, and CENTRAL databases.All studies demonstrated significant increases in mean arterial pressure (MAP) and systemic vascular resistance during vasopressor infusion. The calculated mean systemic filling pressure analogue increased (16 ±â€Š3.3 mmHg to 18 ±â€Š3.4 mmHg; P = 0.02) by vasopressors with variable effects on central venous pressure and the pump efficiency of the heart leading to heterogenous changes in cardiac output. Changes in the pressure gradient for venous return and cardiac output, scaled by the change in MAP, were positively correlated (r2 = 0.88, P < 0.001). Changes in the mean systemic filling pressure analogue and heart pump efficiency were negatively correlated (r2 = 0.57, P < 0.001) while no correlation was found between changes in MAP and heart pump efficiency.We conclude that hemodynamic changes induced by vasopressor therapy are inadequately represented by the change in MAP alone despite its common use as a clinical endpoint. The more comprehensive analysis applied in this review illustrates how vasopressor administration may be optimized.


Assuntos
Débito Cardíaco/efeitos dos fármacos , Choque/fisiopatologia , Resistência Vascular/efeitos dos fármacos , Vasoconstritores/farmacologia , Pressão Venosa/efeitos dos fármacos , Adulto , Pressão Arterial/efeitos dos fármacos , Humanos , Choque/tratamento farmacológico
16.
Circ Heart Fail ; 14(3): e007351, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33663236

RESUMO

BACKGROUND: New heart failure therapies that safely augment cardiac contractility and output are needed. Previous apelin peptide studies have highlighted the potential for APJ (apelin receptor) agonism to enhance cardiac function in heart failure. However, apelin's short half-life limits its therapeutic utility. Here, we describe the preclinical characterization of a novel, orally bioavailable APJ agonist, BMS-986224. METHODS: BMS-986224 pharmacology was compared with (Pyr1) apelin-13 using radio ligand binding and signaling pathway assays downstream of APJ (cAMP, phosphorylated ERK [extracellular signal-regulated kinase], bioluminescence resonance energy transfer-based G-protein assays, ß-arrestin recruitment, and receptor internalization). Acute effects on cardiac function were studied in anesthetized instrumented rats. Chronic effects of BMS-986224 were assessed echocardiographically in the RHR (renal hypertensive rat) model of cardiac hypertrophy and decreased cardiac output. RESULTS: BMS-986224 was a potent (Kd=0.3 nmol/L) and selective APJ agonist, exhibiting similar receptor binding and signaling profile to (Pyr1) apelin-13. G-protein signaling assays in human embryonic kidney 293 cells and human cardiomyocytes confirmed this and demonstrated a lack of signaling bias relative to (Pyr1) apelin-13. In anesthetized instrumented rats, short-term BMS-986224 infusion increased cardiac output (10%-15%) without affecting heart rate, which was similar to (Pyr1) apelin-13 but differentiated from dobutamine. Subcutaneous and oral BMS-986224 administration in the RHR model increased stroke volume and cardiac output to levels seen in healthy animals but without preventing cardiac hypertrophy and fibrosis, effects differentiated from enalapril. CONCLUSIONS: We identify a novel, potent, and orally bioavailable nonpeptidic APJ agonist that closely recapitulates the signaling properties of (Pyr1) apelin-13. We show that oral APJ agonist administration induces a sustained increase in cardiac output in the cardiac disease setting and exhibits a differentiated profile from the renin-angiotensin system inhibitor enalapril, supporting further clinical evaluation of BMS-986224 in heart failure.


Assuntos
Receptores de Apelina/agonistas , Débito Cardíaco/efeitos dos fármacos , Insuficiência Cardíaca/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Volume Sistólico/efeitos dos fármacos , Animais , Técnicas de Transferência de Energia por Ressonância de Bioluminescência , Células CHO , Cricetulus , Cães , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Haplorrinos , Humanos , Técnicas In Vitro , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação , Ensaio Radioligante , Ratos , Trítio , Pressão Ventricular/efeitos dos fármacos , beta-Arrestinas/efeitos dos fármacos , beta-Arrestinas/metabolismo
17.
Medicine (Baltimore) ; 100(6): e23570, 2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33578509

RESUMO

ABSTRACT: The Gas Man simulation software provides an opportunity to teach, understand and examine the pharmacokinetics of volatile anesthetics. The primary aim of this study was to investigate the accuracy of a cardiac output and alveolar ventilation matched Gas Man model and to compare its predictive performance with the standard pharmacokinetic model using patient data.Therefore, patient data from volatile anesthesia were successively compared to simulated administration of desflurane and sevoflurane for the standard and a parameter-matched simulation model with modified alveolar ventilation and cardiac output. We calculated the root-mean-square deviation (RMSD) between measured and calculated induction, maintenance and elimination and the expiratory decrement times during emergence and recovery for the standard and the parameter-matched model.During induction, RMSDs for the standard Gas Man simulation model were higher than for the parameter-matched Gas Man simulation model [induction (desflurane), standard: 1.8 (0.4) % Atm, parameter-matched: 0.9 (0.5) % Atm., P = .001; induction (sevoflurane), standard: 1.2 (0.9) % Atm, parameter-matched: 0.4 (0.4) % Atm, P = .029]. During elimination, RMSDs for the standard Gas Man simulation model were higher than for the parameter-matched Gas Man simulation model [elimination (desflurane), standard: 0.7 (0.6) % Atm, parameter-matched: 0.2 (0.2) % Atm, P = .001; elimination (sevoflurane), standard: 0.7 (0.5) % Atm, parameter-matched: 0.2 (0.2) % Atm, P = .008]. The RMSDs during the maintenance of anesthesia and the expiratory decrement times during emergence and recovery showed no significant differences between the patient and simulated data for both simulation models.Gas Man simulation software predicts expiratory concentrations of desflurane and sevoflurane in humans with good accuracy, especially when compared to models for intravenous anesthetics. Enhancing the standard model by ventilation and hemodynamic input variables increases the predictive performance of the simulation model. In most patients and clinical scenarios, the predictive performance of the standard Gas Man simulation model will be high enough to estimate pharmacokinetics of desflurane and sevoflurane with appropriate accuracy.


Assuntos
Débito Cardíaco/efeitos dos fármacos , Desflurano/farmacocinética , Expiração/fisiologia , Ventilação Pulmonar/fisiologia , Sevoflurano/farmacocinética , Adulto , Idoso , Algoritmos , Anestésicos Inalatórios/administração & dosagem , Anestésicos Inalatórios/farmacocinética , Débito Cardíaco/fisiologia , Ensaios Clínicos como Assunto , Simulação por Computador/estatística & dados numéricos , Desflurano/administração & dosagem , Quimioterapia Combinada , Feminino , Humanos , Pulmão/metabolismo , Pulmão/fisiologia , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/fisiologia , Sevoflurano/administração & dosagem
18.
Physiol Rep ; 9(3): e14753, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33587331

RESUMO

Chlorisondamine (CSD) has been used to assess the neurogenic contribution to blood pressure (BP) and vasomotor sympathetic tone in animal models. It is assumed that the reduction in BP following CSD administration is associated to decreases in cardiac output (CO) and peripheral resistance, reflecting cardiac and vasomotor sympathetic tone, respectively. Surprisingly, this has not been characterized experimentally in mice, despite the extensive use of this animal model in cardiovascular research. We hypothesize that a specific dose of CSD can selectively block the sympathetic vasomotor tone. To test this hypothesis, we evaluated the effects of different doses of CSD (intraperitoneal) on BP and heart rate (HR) using telemetry, and on CO using echocardiography. BP and HR in normotensive C57Bl/6J mice reduced to a similar extent by all CSD doses tested (1-6 mg/kg). CSD at 6 mg/kg also reduced CO without affecting left ventricular stroke volume or fractional shortening. On the other hand, lower doses of CSD (1 and 2 mg/kg) produced significantly larger BP and HR reductions in DOCA-salt-induced hypertensive mice, indicating a greater neurogenic BP response. In addition, all doses of CSD reduced CO in hypertensive mice. Our data suggest that the BP response to CSD in mice likely reflects reduced CO and vasomotor sympathetic tone. We conclude that CSD can be used to assess the neurogenic contribution to BP in mice but may not be appropriate for specifically estimating vasomotor sympathetic tone.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Sistema Cardiovascular/inervação , Clorisondamina/farmacologia , Hipertensão/fisiopatologia , Sistema Nervoso Simpático/efeitos dos fármacos , Simpatolíticos/farmacologia , Animais , Débito Cardíaco/efeitos dos fármacos , Acetato de Desoxicorticosterona , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/etiologia , Masculino , Camundongos Endogâmicos C57BL , Cloreto de Sódio na Dieta , Sistema Nervoso Simpático/fisiopatologia , Sistema Vasomotor/efeitos dos fármacos , Sistema Vasomotor/fisiopatologia
19.
Pharmacol Res Perspect ; 9(1): e00714, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33507583

RESUMO

Coronary microembolization (CME)-induced inflammation and cardiomyocyte apoptosis are two key factors contributing to CME-induced myocardial dysfunction. High-mobility group box-1 (HMGB1) plays essential role in progression of CME-induced injury and inhibition of HMGB1 has been shown to be protective. In present study, the potential effects of glycyrrhizin, a HMGB1 inhibitor, on CME-induced myocardial dysfunction are evaluated. Using a rat model of CME, we administrated glycyrrhizin in rats prior to CME induction. The level of HMGB1, TNF-α, iNOS, IL-6, IL-1ß, cleaved caspase-3, Bax, and Bcl-2 were measured. The serum level of cardiac troponin I, creatine kinase, was detected. The cardiac function and cardiomyocyte apoptosis were evaluated. The activation of TLR4/NF-κB signaling pathway was analyzed. Glycyrrhizin prevented CME-induced production of HMGB1, TNF-α, iNOS, IL-6, and IL-1ß. Glycyrrhizin inhibited CME-induced cardiomyocyte apoptosis and the expression of cleaved caspase-3 and Bax, while enhanced the expression of Bcl-2. Glycyrrhizin decreased cardiac troponin I and creatine kinase levels and improved cardiac function. Glycyrrhizin prevented the activation of HMGB1/TLR4/NF-κB signaling pathway. Glycyrrhizin ameliorated myocardial dysfunction in CME rats by preventing inflammation and apoptosis of cardiomyocytes.


Assuntos
Anti-Inflamatórios/uso terapêutico , Cardiotônicos/uso terapêutico , Embolia/tratamento farmacológico , Ácido Glicirrízico/uso terapêutico , Proteína HMGB1/antagonistas & inibidores , Disfunção Ventricular Esquerda/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Débito Cardíaco/efeitos dos fármacos , Cardiotônicos/farmacologia , Vasos Coronários , Citocinas/genética , Citocinas/metabolismo , Embolia/genética , Embolia/metabolismo , Ácido Glicirrízico/farmacologia , Proteína HMGB1/sangue , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Masculino , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo
20.
Crit Care ; 25(1): 21, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33413583

RESUMO

BACKGROUND: Several studies have shown that heart rate control with selective beta-1 blockers in septic shock is safe. In these trials, esmolol was administered 24 h after onset of septic shock in patients who remained tachycardic. While an earlier use of beta-blockers might be beneficial, such use remains challenging due to the difficulty in distinguishing between compensatory and non-compensatory tachycardia. Therefore, the Esmosepsis study was designed to study the effects of esmolol aimed at reducing the heart rate by 20% after the initial resuscitation process in hyperkinetic septic shock patients on (1) cardiac index and (2) systemic and regional hemodynamics as well as inflammatory patterns. METHODS: Nine consecutive stabilized tachycardic hyperkinetic septic shock patients treated with norepinephrine for a minimum of 6 h were included. Esmolol was infused during 6 h in order to decrease the heart rate by 20%. The following data were recorded at hours H0 (before esmolol administration), H1-H6 (esmolol administration) and 1 h after esmolol cessation (H7): systolic arterial pressure, diastolic arterial pressure, mean arterial pressure, central venous pressure, heart rate, PICCO transpulmonary thermodilution, sublingual and musculo-cutaneous microcirculation, indocyanine green clearance and echocardiographic parameters, diuresis, lactate, and arterial and venous blood gases. RESULTS: Esmolol was infused 9 (6.4-11.6) hours after norepinephrine introduction. Esmolol was ceased early in 3 out of 9 patients due to a marked increase in norepinephrine requirement associated with a picture of persistent cardiac failure at the lowest esmolol dose. For the global group, during esmolol infusion, norepinephrine infusion increased from 0.49 (0.34-0.83) to 0.78 (0.3-1.11) µg/min/kg. The use of esmolol was associated with a significant decrease in heart rate from 115 (110-125) to 100 (92-103) beats/min and a decrease in cardiac index from 4.2 (3.1-4.4) to 2.9 (2.5-3.7) l/min/m-2. Indexed stroke volume remained unchanged. Cardiac function index and global ejection fraction also markedly decreased. Using echocardiography, systolic, diastolic as well as left and right ventricular function parameters worsened. After esmolol cessation, all parameters returned to baseline values. Lactate and microcirculatory parameters did not change while the majority of pro-inflammatory proteins decreased in all patients. CONCLUSION: In the very early phase of septic shock, heart rate reduction using fast esmolol titration is associated with an increased risk of hypotension and decreased cardiac index despite maintained adequate tissue perfusion (NCT02068287).


Assuntos
Hemodinâmica/efeitos dos fármacos , Propanolaminas/farmacologia , Choque Séptico/tratamento farmacológico , Fatores de Tempo , Adulto , Idoso , Idoso de 80 Anos ou mais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Débito Cardíaco/efeitos dos fármacos , Ecocardiografia/métodos , Feminino , França , Frequência Cardíaca/efeitos dos fármacos , Humanos , Verde de Indocianina/análise , Masculino , Pessoa de Meia-Idade , Monitorização Fisiológica/métodos , Norepinefrina/administração & dosagem , Norepinefrina/classificação , Projetos Piloto , Propanolaminas/uso terapêutico , Choque Séptico/fisiopatologia , Espectrofotometria Infravermelho/métodos , Vasoconstritores/administração & dosagem , Vasoconstritores/classificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...