Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.654
Filter
1.
Trials ; 25(1): 558, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39180100

ABSTRACT

BACKGROUND: Tracheal intubation may cause significant hemodynamic responses. Many drugs have been shown to be effective in modifying these cardiovascular responses, including remifentanil, fentanyl, sufentanil, and alfentanil. However, the 90% effect-site concentration (EC90) of remifentanil required to control cardiovascular responses to tracheal intubation when combined with ciprofol remains unclear. The purpose of this study was to determine the EC90 of remifentanil inhibiting cardiovascular responses to tracheal intubation during anesthesia induction with ciprofol using biased-coin design up-and-down sequential method (BC-UDM). METHODS: This is a prospective sequential allocation dose-finding study. American Society of Anesthesiologists physical status (ASA) I-II elective surgical patients receiving target-controlled infusion (TCI) of remifentanil effect-site concentration (Ce), followed by ciprofol and rocuronium for anesthesia, were enrolled. The cardiovascular response to tracheal intubation was defined as positive when mean arterial pressure (MAP) or heart rate (HR) is 15% higher than the baseline value. Using the BC-UDM, the Ce of remifentanil was determined based on the cardiovascular response to tracheal intubation of the previous patient. The EC90 and 90% confidence intervals (90% CIs) were estimated by R-Foundation centered isotonic regression and the pooled adjacent violators algorithm with bootstrapping. DISCUSSION: The results of this study sought to demonstrate EC90 of remifentanil blunting sympathetic responses to tracheal intubation during anesthesia index (Ai)-guided ciprofol anesthesia using BCD-UDM. It may help to minimize the cardiovascular responses to tracheal intubation. TRIAL REGISTRATION: Chinese Clinical Trial Registry ChiCTR2300078275. Registered on December 3, 2023.


Subject(s)
Heart Rate , Intubation, Intratracheal , Remifentanil , Humans , Remifentanil/administration & dosage , Intubation, Intratracheal/methods , Prospective Studies , Heart Rate/drug effects , Male , Anesthetics, Intravenous/administration & dosage , Female , Adult , Dose-Response Relationship, Drug , Middle Aged , Arterial Pressure/drug effects , Hemodynamics/drug effects , Rocuronium/administration & dosage , Analgesics, Opioid/administration & dosage
2.
BMJ Open ; 14(8): e086388, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39117412

ABSTRACT

INTRODUCTION: The dynamic arterial elastance (EaDyn), calculated as pulse pressure variation divided by stroke volume variation, has been studied as a predictor of vasopressor weaning. However, its potential as a haemodynamic tool for tapering off vasopressors in patients with sepsis remains unexplored. Therefore, our study aimed to assess whether using EaDyn for weaning vasopressor support could reduce the duration of vasopressor support in patients with sepsis. METHODS AND ANALYSIS: This pragmatic single-centre controlled clinical trial will take place at Fundación Santa Fe de Bogotá, Colombia. Adult patients diagnosed with septic shock according to the sepsis-3 criteria and a Sequential Organ Failure Assessment score ≥4 will be included. A total of 114 patients (57 per group) will undergo conventional critical care monitoring, and the weaning of vasopressor support will be initiated based on the EaDyn or mean arterial pressure (MAP), depending on the assigned group. EaDyn will be estimated based on the measurements obtained from a PiCCO device connected to a PulsioFlex Monitoring Platform (PULSION Medical Systems SE, Feldkirchen, Germany). Our primary outcome is the difference in vasopressor support duration between the EaDyn and MAP groups.Participants and statisticians performing the statistical analysis will be blinded to the group allocation. Dependent and independent variables will be analysed through univariate and multivariate statistical tests. Since we will perform three repeated measurements for analysis, we will implement a Bonferroni post hoc correction. Additionally, Cox regression and Kaplan-Meier analyses will be conducted to address objectives related to time. ETHICS AND DISSEMINATION: This study was approved by the Ethics Committee at Fundación Santa Fe de Bogotá (CCEI-16026-2024). Written informed consent will be obtained from all participants. The results will be disseminated through publication in peer-reviewed journals and presentations at national and international events. TRIAL REGISTRATION NUMBER: NCT06118775.


Subject(s)
Shock, Septic , Vasoconstrictor Agents , Humans , Shock, Septic/drug therapy , Shock, Septic/physiopathology , Vasoconstrictor Agents/therapeutic use , Randomized Controlled Trials as Topic , Stroke Volume , Male , Colombia , Female , Arterial Pressure/drug effects , Critical Care/methods , Adult
3.
Crit Care Explor ; 6(8): e1141, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39120069

ABSTRACT

OBJECTIVE: Mean arterial pressure (MAP) plays a significant role in regulating tissue perfusion and urine output (UO). The optimal MAP target in critically ill patients remains a subject of debate. We aimed to explore the relationship between MAP and UO. DESIGN: A retrospective observational study. SETTING: A general ICU in a tertiary medical center. PATIENTS: All critically ill patients admitted to the ICU for more than 10 hours. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: MAP values and hourly UO were collected in 5,207 patients. MAP levels were categorized into 10 groups of 5 mm Hg (from MAP < 60 mm Hg to MAP > 100 mg Hg), and 656,423 coupled hourly mean MAP and UO measurements were analyzed. Additionally, we compared the UO of individual patients in each MAP group with or without norepinephrine (NE) support or diuretics, as well as in patients with acute kidney injury (AKI).Hourly UO rose incrementally between MAP values of 65-100 mm Hg. Among 2,226 patients treated with NE infusion, mean UO was significantly lower in the MAP less than 60 mm Hg group (53.4 mL/hr; 95% CI, 49.3-57.5) compared with all other groups (p < 0.001), but no differences were found between groups of 75 less than or equal to MAP. Among 2500 patients with AKI, there was a linear increase in average UO from the MAP less than 60 mm Hg group (57.1 mL/hr; 95% CI, 54.2-60.0) to the group with MAP greater than or equal to 100 mm Hg (89.4 mL/hr; 95% CI, 85.7-93.1). When MAP was greater than or equal to 65 mm Hg, we observed a statistically significant trend of increased UO in periods without NE infusion. CONCLUSIONS: Our analysis revealed a linear correlation between MAP and UO within the range of 65-100 mm Hg, also observed in the subgroup of patients treated with NE or diuretics and in those with AKI. These findings highlight the importance of tissue perfusion to the maintenance of diuresis and achieving adequate fluid balance in critically ill patients.


Subject(s)
Arterial Pressure , Critical Illness , Intensive Care Units , Humans , Retrospective Studies , Male , Female , Middle Aged , Arterial Pressure/drug effects , Arterial Pressure/physiology , Aged , Acute Kidney Injury/physiopathology , Acute Kidney Injury/urine , Acute Kidney Injury/diagnosis , Norepinephrine/urine , Urination/drug effects , Urination/physiology
4.
PLoS One ; 19(7): e0307407, 2024.
Article in English | MEDLINE | ID: mdl-39024364

ABSTRACT

One of the most important components of sepsis management is hemodynamic restoration. If the target mean arterial pressure (MAP) is not obtained, the first recommendation is for volume expansion, and the second is for norepinephrine (NE). We describe the methodology of a randomized multicenter trial aiming to assess the hypothesis that low-dose NE given early in adult patients with sepsis will provide better control of shock within 6 hours from therapy starting compared to standard care. This trial includes ICU septic patients in whom MAP decrease below 65 mmHg to be randomized into 2 groups: early NE-group versus standard care-group. The patient's attending clinician will determine how much volume expansion is necessary to meet the target of a MAP > 65 mm Hg. If this target not achieved, after at least 30 ml/kg and guided by the available indices of fluid responsiveness, NE will be used in a usual way. The latter must follow a consensual schedule elaborated by the investigating centers. Parameters to be taken at inclusion and at H6 are: lactates, cardiac ultrasound parameters (stroke volume (SV), cardiac output (CO), E/E' ratio), and P/F ratio. MAP and diuresis are recorded hourly. Our primary outcome is the shock control defined as a composite criterion (MAP > 65 mm Hg for 2 consecutive measurements and urinary output > 0.5 ml/kg/h for 2 consecutive hours) within 6 hours. Secondary outcomes: Decrease in serum lactate> 10% from baseline within 6 hours, the received fluid volume within 6 hours, variation of CO and E/E', and 28 days-Mortality. The study is ongoing and aims to include at least 100 patients per arm. This study is likely to contribute to support the indication of early initiation of NE with the aim to restrict fluid intake in septic patients. (ClinicalTrials.gov ID: NCT05836272).


Subject(s)
Norepinephrine , Sepsis , Humans , Norepinephrine/administration & dosage , Sepsis/drug therapy , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/therapeutic use , Adult , Hemodynamics , Cardiac Output , Arterial Pressure/drug effects , Male , Female
5.
Cardiovasc Toxicol ; 24(9): 843-851, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38963633

ABSTRACT

The contribution of air pollution-induced cardiopulmonary damage on the development of hypertensive disorders of pregnancy and other adverse outcomes of pregnancy has gained increased attention as epidemiological data continue to highlight spatiotemporal pregnancy trends related to air pollution exposure. However clinical mechanistic data surrounding gestational complications remain sparse, necessitating the need for the use of animal models to study these types of complications of pregnancy. The current study seeks to examine the real-time effects of mid-gestational ozone exposure on maternal blood pressure and body temperature through the use of radiotelemetry in a rat model. The exposure resulted in acute depression of heart rate and core body temperature as compared to control animals. Ozone-exposed animals also presented with a slight but significant increase in arterial blood pressure which was perpetuated until term. The data presented here illustrates the feasibility of murine models to assess cardiovascular complications caused by inhaled toxicants during the window of pregnancy.


Subject(s)
Disease Models, Animal , Heart Rate , Hypertension, Pregnancy-Induced , Ozone , Animals , Pregnancy , Female , Ozone/toxicity , Heart Rate/drug effects , Hypertension, Pregnancy-Induced/chemically induced , Hypertension, Pregnancy-Induced/physiopathology , Time Factors , Arterial Pressure/drug effects , Rats, Sprague-Dawley , Air Pollutants/toxicity , Gestational Age , Inhalation Exposure/adverse effects , Telemetry , Rats , Prenatal Exposure Delayed Effects/chemically induced , Maternal Exposure/adverse effects
6.
J Cardiovasc Pharmacol ; 84(3): 356-369, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39027975

ABSTRACT

ABSTRACT: Angiomotin-like 2 (AMOTL2) is related to numerous physiological and pathological conditions by affecting signal transduction. However, whether AMOTL2 is linked to pulmonary arterial hypertension (PAH) has not been addressed. This work aimed to investigate the potential role of AMOTL2 in PAH. A decrease in AMOTL2 abundance was observed in the lungs of PAH rats. The upregulation of AMOTL2 significantly decreased right ventricle systolic pressure and right ventricular hypertrophy in PAH rats. Overexpression of AMOTL2 also led to a noteworthy decrease in vascular wall thickness, pulmonary artery area, and collagen deposition in rats with PAH. AMOTL2 was downregulated in hypoxia-stimulated pulmonary arterial smooth muscle cells (PASMCs). Moreover, AMOTL2 overexpression impeded hypoxia-evoked proliferation, migration, and phenotypic transformation in rat PASMCs. Mechanistic investigation revealed that Yes-associated protein 1 (YAP1) activation in PAH rats or hypoxia-stimulated PASMCs was markedly inhibited by AMOTL2 overexpression, which was associated with increased large tumor suppressor 1/2 phosphorylation. The inhibition of large tumor suppressor 1/2 reversed the AMOTL2-mediated inactivation of YAP1. Restoring the activity of YAP1 reversed the inhibitory effect of AMOTL2 on hypoxia-evoked proliferation, migration, and phenotypic transformation of PASMCs. Collectively, these results suggest that AMOTL2 can ameliorate PAH in a rat model by interfering with pulmonary arterial remodeling via the inactivation of YAP1 signaling. Our work indicates that AMOTL2 may be a candidate target for novel drug development for the treatment of PAH.


Subject(s)
Angiomotins , Cell Proliferation , Disease Models, Animal , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Pulmonary Arterial Hypertension , Pulmonary Artery , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation , Vascular Remodeling , YAP-Signaling Proteins , Animals , YAP-Signaling Proteins/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/drug effects , Vascular Remodeling/drug effects , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Cells, Cultured , Cell Proliferation/drug effects , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/pathology , Cell Movement/drug effects , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/pathology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Ventricular Function, Right/drug effects , Arterial Pressure/drug effects , Phenotype , Rats , Phosphorylation
7.
Am J Physiol Renal Physiol ; 327(2): F199-F207, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38841747

ABSTRACT

Chronic kidney disease is the loss of renal function that can occur from aging or through a myriad of other disease states. Rising serum concentrations of kynurenine, a tryptophan metabolite, have been shown to correlate with increasing severity of chronic kidney disease. This study used chronic intravenous infusion in conscious male Sprague-Dawley rats to test the hypothesis that kynurenine can induce renal damage and promote alterations in blood pressure, heart rate, and decreased renal function. We found that kynurenine infusion increased mean arterial pressure, increased the maximum and minimum range of heart rate, decreased glomerular filtration rate, and induced kidney damage in a dose-dependent manner. This study shows that kynurenine infusion can promote kidney disease in healthy, young rats, implying that the increase in kynurenine levels associated with chronic kidney disease may establish a feed-forward mechanism that exacerbates the loss of renal function.NEW & NOTEWORTHY In humans, an elevated serum concentration of kynurenine has long been associated with negative outcomes in various disease states as well as in aging. However, it has been unknown whether these increased kynurenine levels are mediating the disorders or simply associated with them. This study shows that chronically infusing kynurenine can contribute to the development of hypertension and kidney impairment. The mechanism of this action remains to be determined in future studies.


Subject(s)
Arterial Pressure , Glomerular Filtration Rate , Kidney , Kynurenine , Rats, Sprague-Dawley , Tryptophan , Animals , Kynurenine/blood , Kynurenine/metabolism , Male , Arterial Pressure/drug effects , Tryptophan/blood , Tryptophan/metabolism , Glomerular Filtration Rate/drug effects , Kidney/metabolism , Kidney/drug effects , Kidney/physiopathology , Infusions, Intravenous , Heart Rate/drug effects , Rats , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/blood
9.
J Appl Physiol (1985) ; 137(1): 136-144, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38813608

ABSTRACT

Contemporary discussion of the baroreflex includes the efferent vascular-sympathetic and cardiovagal arms. Since sympathetic postganglionic neurons also innervate the left ventricle (LV), it is often assumed that the LV produces a sympathetically mediated increase in contractility during baroreceptor unloading, but this has not been characterized using a load-independent index of contractility. We aimed to determine 1) whether LV contractility increases in response to baroreceptor unloading and 2) whether such increases are mediated via the sympathetic or parasympathetic arm of the autonomic nervous system. Ten male Wistar rats were anesthetized (urethane) and instrumented with arterial and LV pressure-volume catheters to measure mean arterial pressure (MAP) and load-independent LV contractility [maximal rate of increase in pressure adjusted to end-diastolic volume (PAdP/dtmax)], respectively. Rats were placed in a servo-controlled lower-body negative pressure (LBNP) chamber to reduce MAP by 10% for 60 s to mechanically unload baroreceptors under control conditions. LBNP was repeated in each animal following infusions of cardiac autonomic blockers using esmolol (sympathetic), atropine (parasympathetic), and esmolol + atropine. Under control conditions, PAdP/dtmax increased during baroreceptor unloading (26 ± 6 vs. 31 ± 9 mmHg·s-1·µL-1, P = 0.031). During esmolol, there was no increase in LV contractility during baroreceptor unloading (11 ± 2 vs. 12 ± 2, P = 0.125); however, during atropine, there was an increase in LV contractility during baroreceptor unloading (26 ± 6 vs. 31 ± 9, P = 0.019). During combined esmolol and atropine, there was a small increase in contractility versus control (13 ± 3 vs. 15 ± 4, P = 0.046). Our results demonstrate that, in anesthetized rats, LV contractility increases in response to baroreceptor unloading, which is largely sympathetically mediated.NEW & NOTEWORTHY This study empirically demonstrates a sympathetically mediated increase in LV contractility in response to baroreceptor unloading using a load-independent index of cardiac contractility in the anesthetized rat.


Subject(s)
Baroreflex , Heart Ventricles , Myocardial Contraction , Pressoreceptors , Rats, Wistar , Sympathetic Nervous System , Ventricular Function, Left , Animals , Male , Myocardial Contraction/physiology , Myocardial Contraction/drug effects , Rats , Pressoreceptors/physiology , Pressoreceptors/drug effects , Baroreflex/physiology , Baroreflex/drug effects , Sympathetic Nervous System/physiology , Sympathetic Nervous System/drug effects , Heart Ventricles/drug effects , Ventricular Function, Left/physiology , Ventricular Function, Left/drug effects , Blood Pressure/physiology , Blood Pressure/drug effects , Arterial Pressure/physiology , Arterial Pressure/drug effects , Atropine/pharmacology , Anesthesia , Propanolamines
10.
Int J Chron Obstruct Pulmon Dis ; 19: 1105-1121, 2024.
Article in English | MEDLINE | ID: mdl-38803412

ABSTRACT

Purpose: This phase 1 study (NCT04370873) evaluated safety and pharmacokinetics/pharmacodynamics (PK/PD) of MK-5475 in participants with pulmonary hypertension associated with COPD (PH-COPD). Methods: Eligible participants were 40-80 years old with COPD (FEV1/FVC <0.7; FEV1 >30% predicted) and PH (mean pulmonary arterial pressure ≥25 mmHg). Participants were randomized 2:1 to MK-5475 or placebo via dry-powder inhaler once daily for 7 days in Part 1 (360 µg) or 28 days in Part 2 (380 µg). Safety was assessed by adverse events (AEs) and arterial blood oxygenation. Part-2 participants had pulmonary vascular resistance (PVR; primary PD endpoint) and pulmonary blood volume (PBV; secondary PD endpoint) measured at baseline and Day 28. A non-informative prior was used to calculate posterior probability (PP) that the between-group difference (MK-5475 - placebo) in mean percent reduction from baseline in PVR was less than -15%. Results: Nine participants were randomized in Part 1, and 14 participants in Part 2. Median age of participants (86.4% male) was 68.5 years (41-77 years); 95.5% had moderate-to-severe COPD. Incidences of AEs were comparable between MK-5475 and placebo: overall (5/14 [36%] versus 5/8 [63%]), drug-related (1/14 [7%] versus 2/8 [25%]), and serious (1/14 [7%] versus 1/8 [13%]). MK-5475 caused no meaningful changes in arterial blood oxygenation or PBV. MK-5475 versus placebo led to numerical improvements from baseline in PVR (-21.2% [95% CI: -35.4, -7.0] versus -5.4% [95% CI: -83.7, 72.9]), with between-group difference in PVR less than -15% and calculated PP of 51%. Conclusion: The favorable safety profile and numerical reductions in PVR observed support further clinical development of inhaled MK-5475 for PH-COPD treatment.


Subject(s)
Hypertension, Pulmonary , Lung , Pulmonary Disease, Chronic Obstructive , Humans , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/physiopathology , Male , Aged , Administration, Inhalation , Female , Middle Aged , Treatment Outcome , Double-Blind Method , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/diagnosis , Adult , Lung/physiopathology , Lung/drug effects , Aged, 80 and over , Soluble Guanylyl Cyclase/metabolism , Dry Powder Inhalers , Time Factors , Forced Expiratory Volume , Enzyme Activators/administration & dosage , Enzyme Activators/adverse effects , Enzyme Activators/pharmacokinetics , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/adverse effects , Antihypertensive Agents/pharmacokinetics , Arterial Pressure/drug effects , Vital Capacity
11.
Clin Exp Rheumatol ; 42(8): 1615-1622, 2024 08.
Article in English | MEDLINE | ID: mdl-38819960

ABSTRACT

OBJECTIVES: Bosentan is a dual endothelin receptor antagonist approved for the treatment of SSc digital ulcers (DU) and pulmonary arterial hypertension (PAH). Systolic pulmonary arterial pressure (sPAP) is a relevant parameter for the follow-up and prognosis of SSc-PAH. The therapeutic magnitude of bosentan in SSc-PAH is not fully understood, thus we aim to establish the degree of sPAP reduction in bosentan treated SSc-PAH patients. METHODS: We performed a systematic literature review in three databases from January 2000 to June 2023, involving sPAP measurement at transthoracic echocardiography of SSc patients before and after starting bosentan. Following the study quality assessment and data extraction, we performed random-effects meta-analysis and Egger's test for publication bias. Stratified analysis was performed for mono-/combination therapy, follow up duration (≤1 year), indication for bosentan therapy (PAH or DU/mixed). RESULTS: In the 11 selected manuscripts, sPAP mean difference before and after bosentan therapy was - 5.63mmHg (CI95% -9.79 to -1.48, p=0.0078). In stratified analysis, sPAP mean was significantly different before and after bosentan therapy only for studies considering < 1 year of follow-up (p=0.0020), monotherapy (p=0.0140) and the strict indication for PAH (p=0.0002). CONCLUSIONS: Bosentan significantly decreases sPAP, a relevant prognostic marker, especially in overt SSc-PAH. However, bosentan did not decrease sPAP when started for DU/mixed indication nor for follow-up>1 year. The burden of publication bias was significant. Therefore, further studies are required to assess bosentan's haemodynamic effect in high-risk patients for SSc-PAH.


Subject(s)
Antihypertensive Agents , Arterial Pressure , Bosentan , Endothelin Receptor Antagonists , Hypertension, Pulmonary , Pulmonary Artery , Scleroderma, Systemic , Bosentan/therapeutic use , Humans , Scleroderma, Systemic/complications , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/physiopathology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/diagnostic imaging , Antihypertensive Agents/therapeutic use , Endothelin Receptor Antagonists/therapeutic use , Arterial Pressure/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Artery/diagnostic imaging , Echocardiography , Treatment Outcome , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/diagnostic imaging
12.
Am J Physiol Heart Circ Physiol ; 327(1): H000, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38819383

ABSTRACT

Nitric oxide (NO) inhalation improves pulmonary hemodynamics in participants with pulmonary arterial hypertension (PAH). Although it can reduce pulmonary vascular resistance (PVR) in PAH, its impact on the dynamic mechanics of pulmonary arteries and its potential difference between control and participants with PAH remain unclear. PA impedance provides a comprehensive description of PA mechanics. With an arterial model, PA impedance can be parameterized into peripheral pulmonary resistance (Rp), arterial compliance (Cp), characteristic impedance of the proximal arteries (Zc), and transmission time from the main PA to the reflection site. This study investigated the effects of inhaled NO on PA impedance and its associated parameters in control and monocrotaline-induced pulmonary arterial hypertension (MCT-PAH) male rats (6/group). Measurements were obtained at baseline and during NO inhalation at 40 and 80 ppm. In both groups, NO inhalation decreased PVR and increased the left atrial pressure. Notably, its impact on PA impedance was frequency dependent, as revealed by reduced PA impedance modulus in the low-frequency range below 10 Hz, with little effect on the high-frequency range. Furthermore, NO inhalation attenuated Rp, increased Cp, and prolonged transmission time without affecting Zc. It reduced Rp more pronouncedly in MCT-PAH rats, whereas it increased Cp and delayed transmission time more effectively in control rats. In conclusion, the therapeutic effects of inhaled NO on PA impedance were frequency dependent and may differ between the control and MCT-PAH groups, suggesting that the effect on the mechanics differs depending on the pathological state.NEW & NOTEWORTHY Nitric oxide inhalation decreased pulmonary arterial impedance in the low-frequency range (<10 Hz) with little impact on the high-frequency range. It reduced peripheral pulmonary resistance more pronouncedly in pulmonary hypertension rats, whereas it increased arterial compliance and transmission time in control rats. Its effect on the mechanics of the pulmonary arteries may differ depending on the pathological status.


Subject(s)
Nitric Oxide , Pulmonary Artery , Vascular Resistance , Animals , Male , Nitric Oxide/metabolism , Pulmonary Artery/physiopathology , Pulmonary Artery/drug effects , Administration, Inhalation , Vascular Resistance/drug effects , Monocrotaline , Rats , Rats, Sprague-Dawley , Disease Models, Animal , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/chemically induced , Arterial Pressure/drug effects
14.
Braz J Anesthesiol ; 74(4): 844503, 2024.
Article in English | MEDLINE | ID: mdl-38641324

ABSTRACT

BACKGROUND: The incidence of arterial hypotension during induction of general anesthesia is influenced by the method of propofol administration, but there is a dearth of randomized clinical trials comparing bolus injection and target-controlled infusion in relation to arterial hypotension. This study seeks to compare the incidence of arterial hypotension between these two methods of propofol administration. METHODS: This prospective, randomized, single-center, non-blinded study included 60 patients (aged 35 to 55 years), classified as ASA physical status I or II, who were undergoing non-cardiac surgeries. They were randomly allocated using a computer to two groups based on the method of propofol administration during the induction of general anesthesia: the Target Group, receiving target-controlled infusion at 4 µg.mL-1, and the Bolus Group, receiving a bolus infusion of 2 mg.kg-1. Both groups also received midazolam 2 mg, fentanyl 3 µg.kg-1, and rocuronium 0.6 mg.kg-1. Over the first 10 minutes of anesthesia induction, Mean Arterial Pressure (MAP), Heart Rate (HR), level of Consciousness (qCON), and Suppression Rate (SR) were recorded every 2 minutes. RESULTS: Twenty-seven patients remained in the TCI group, while 28 were in the Bolus group. Repeated measure analysis using mixed-effects models could not reject the null hypothesis for the effect of group-time interactions in MAP (p = 0.85), HR (p = 0.49), SR (p = 0.44), or qCON (p = 0.72). The difference in means for qCON (60.2 for TCI, 50.5 for bolus, p < 0.001), MAP (90.3 for TCI, 86.2 for bolus, p < 0.006), HR (76.2 for TCI, 76.9 for bolus, p = 0.93), and SR (0.01 for TCI, 5.5 for bolus, p < 0.001), irrespective of time (whole period means), revealed some significant differences. CONCLUSION: Patients who received propofol bolus injection exhibited a lower mean arterial pressure, a greater variation in the level of consciousness, and a higher suppression rate compared to those who received it as a target-controlled infusion. However, the interaction effect between groups and time remains inconclusive.


Subject(s)
Anesthesia, General , Anesthetics, Intravenous , Hypotension , Propofol , Humans , Propofol/administration & dosage , Propofol/adverse effects , Adult , Middle Aged , Anesthesia, General/methods , Female , Male , Hypotension/epidemiology , Hypotension/chemically induced , Prospective Studies , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/adverse effects , Infusions, Intravenous , Incidence , Injections, Intravenous , Arterial Pressure/drug effects
15.
Vascul Pharmacol ; 155: 107374, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642596

ABSTRACT

A comprehensive evaluation of risk, using multiple indices, is necessary to provide reliable prognostic information and guide therapy in pulmonary arterial hypertension (PAH). The current ESC/ERS guidelines suggest using a three-strata model for incident (newly diagnosed) patients and a four-strata model for prevalent patients with PAH. The four-strata model serves as a fundamental risk-stratification tool and relies on a minimal dataset of indicators that must be considered during follow-up. Nevertheless, there are still areas of vagueness and ambiguity when classifying and managing patients in the intermediate-risk category. For these patients, considerations should include right heart imaging, hemodynamics, as well as individual factors such as age, sex, genetic profile, disease type, comorbidities, and kidney function. The aim of this report is to present case studies, with a specific focus on patients ultimately classified as intermediate risk. We aim to emphasize the challenges and complexities encountered in the realms of diagnosis, classification, and treatment for these particular patients.


Subject(s)
Antihypertensive Agents , Practice Guidelines as Topic , Pulmonary Arterial Hypertension , Humans , Risk Factors , Risk Assessment , Antihypertensive Agents/therapeutic use , Pulmonary Arterial Hypertension/diagnosis , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/therapy , Pulmonary Arterial Hypertension/epidemiology , Female , Male , Predictive Value of Tests , Evidence-Based Medicine/standards , Treatment Outcome , Middle Aged , Clinical Decision-Making , Pulmonary Artery/physiopathology , Arterial Pressure/drug effects , Decision Support Techniques
16.
J Cardiovasc Pharmacol ; 84(1): 101-109, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38573589

ABSTRACT

ABSTRACT: Myocardial infarction (MI) and pulmonary arterial hypertension (PAH) are 2 prevalent cardiovascular diseases. In both conditions, oxidative stress is associated with a worse prognosis. Pterostilbene (PTE), an antioxidant compound, has been studied as a possible therapy for cardiovascular diseases. This study aims to evaluate the effect of PTE on oxidative stress in the hearts of animals with MI and in the lungs of animals with PAH. Male Wistar rats were used in both models. In the MI model, the experimental groups were sham, MI, and MI + PTE. In the PAH model, the experimental groups were control, PAH, and PAH + PTE. Animals were exposed to MI through surgical ligation of the left coronary artery, or to PAH, by the administration of monocrotaline (60 mg/kg). Seven days after undergoing cardiac injury, the MI + PTE animals were treated with PTE (100 mg/kg day) for 8 days. After this, the heart was collected for molecular analysis. The PAH + PTE animals were treated with PTE (100 mg/kg day) for 14 days, beginning 7 days after PAH induction. After this, the lungs were collected for biochemical evaluation. We found that PTE administration attenuated the decrease in ejection fraction and improved left ventricle end-systolic volume in infarcted animals. In the PAH model, PTE improved pulmonary artery flow and decreased reactive oxygen species levels in the lung. PTE administration promoted protective effects in terms of oxidative stress in 2 experimental models of cardiac diseases: MI and PAH. PTE also improved cardiac function in infarcted rats and pulmonary artery flow in animals with PAH.


Subject(s)
Antioxidants , Disease Models, Animal , Lung , Myocardial Infarction , Myocardium , Oxidative Stress , Pulmonary Arterial Hypertension , Rats, Wistar , Stilbenes , Animals , Oxidative Stress/drug effects , Male , Myocardial Infarction/physiopathology , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Stilbenes/pharmacology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/metabolism , Antioxidants/pharmacology , Myocardium/metabolism , Myocardium/pathology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Artery/metabolism , Ventricular Function, Left/drug effects , Rats , Reactive Oxygen Species/metabolism , Arterial Pressure/drug effects , Monocrotaline
17.
J Clin Anesth ; 95: 111459, 2024 08.
Article in English | MEDLINE | ID: mdl-38599161

ABSTRACT

STUDY OBJECTIVE: Processed electroencephalography (pEEG) may help clinicians optimize depth of general anesthesia. Avoiding excessive depth of anesthesia may reduce intraoperative hypotension and the need for vasopressors. We tested the hypothesis that pEEG-guided - compared to non-pEEG-guided - general anesthesia reduces the amount of norepinephrine needed to keep intraoperative mean arterial pressure above 65 mmHg in patients having vascular surgery. DESIGN: Randomized controlled clinical trial. SETTING: University Medical Center Hamburg-Eppendorf, Hamburg, Germany. PATIENTS: 110 patients having vascular surgery. INTERVENTIONS: pEEG-guided general anesthesia. MEASUREMENTS: Our primary endpoint was the average norepinephrine infusion rate from the beginning of induction of anesthesia until the end of surgery. MAIN RESULT: 96 patients were analyzed. The mean ± standard deviation average norepinephrine infusion rate was 0.08 ± 0.04 µg kg-1 min-1 in patients assigned to pEEG-guided and 0.12 ± 0.09 µg kg-1 min-1 in patients assigned to non-pEEG-guided general anesthesia (mean difference 0.04 µg kg-1 min-1, 95% confidence interval 0.01 to 0.07 µg kg-1 min-1, p = 0.004). Patients assigned to pEEG-guided versus non-pEEG-guided general anesthesia, had a median time-weighted minimum alveolar concentration of 0.7 (0.6, 0.8) versus 0.8 (0.7, 0.8) (p = 0.006) and a median percentage of time Patient State Index was <25 of 12 (1, 41) % versus 23 (3, 49) % (p = 0.279). CONCLUSION: pEEG-guided - compared to non-pEEG-guided - general anesthesia reduced the amount of norepinephrine needed to keep mean arterial pressure above 65 mmHg by about a third in patients having vascular surgery. Whether reduced intraoperative norepinephrine requirements resulting from pEEG-guided general anesthesia translate into improved patient-centered outcomes remains to be determined in larger trials.


Subject(s)
Anesthesia, General , Electroencephalography , Norepinephrine , Vascular Surgical Procedures , Vasoconstrictor Agents , Humans , Anesthesia, General/methods , Norepinephrine/administration & dosage , Male , Female , Middle Aged , Aged , Electroencephalography/drug effects , Vascular Surgical Procedures/adverse effects , Vasoconstrictor Agents/administration & dosage , Hypotension/prevention & control , Arterial Pressure/drug effects , Monitoring, Intraoperative/methods
18.
Am J Physiol Regul Integr Comp Physiol ; 326(6): R599-R608, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38682242

ABSTRACT

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) with orthostatic intolerance (OI) is characterized by neurocognitive deficits perhaps related to upright hypocapnia and loss of cerebral autoregulation (CA). We performed N-back neurocognition testing and calculated the phase synchronization index (PhSI) between arterial pressure (AP) and cerebral blood velocity (CBV) as a time-dependent measurement of cerebral autoregulation in 11 control (mean age = 24.1 yr) and 15 patients with ME/CFS (mean age = 21.8 yr). All patients with ME/CFS had postural tachycardia syndrome (POTS). A 10-min 60° head-up tilt (HUT) significantly increased heart rate (109.4 ± 3.9 vs. 77.2 ± 1.6 beats/min, P < 0.05) and respiratory rate (20.9 ± 1.7 vs. 14.2 ± 1.2 breaths/min, P < 0.05) and decreased end-tidal CO2 (ETCO2; 33.9 ± 1.1 vs. 42.8 ± 1.2 Torr, P < 0.05) in ME/CFS versus control. In ME/CFS, HUT significantly decreased CBV compared with control (-22.5% vs. -8.7%, P < 0.005). To mitigate the orthostatic CBV reduction, we administered supplemental CO2, phenylephrine, and acetazolamide and performed N-back testing supine and during HUT. Only phenylephrine corrected the orthostatic decrease in neurocognition by reverting % correct n = 4 N-back during HUT in ME/CFS similar to control (ME/CFS = 38.5 ± 5.5 vs. ME/CFS + PE= 65.6 ± 5.7 vs. Control 56.9 ± 7.5). HUT in ME/CFS resulted in increased PhSI values indicating decreased CA. Although CO2 and acetazolamide had no effect on PhSI in ME/CFS, phenylephrine caused a significant reduction in PhSI (ME/CFS = 0.80 ± 0.03 vs. ME/CFS + PE= 0.69 ± 0.04, P < 0.05) and improved cerebral autoregulation. Thus, PE improved neurocognitive function in patients with ME/CFS, perhaps related to improved neurovascular coupling, cerebral autoregulation, and maintenance of CBV.NEW & NOTEWORTHY We evaluated cognitive function before and after CO2, acetazolamide, and phenylephrine, which mitigate orthostatic reductions in cerebral blood velocity. Neither CO2 nor acetazolamide affected N-back testing (% correct answers) during an orthostatic challenge. Only phenylephrine improved upright N-back performance in ME/CFS, as it both blocked hyperventilation and increased CO2 significantly compared with those untreated. And only phenylephrine resulted in improved PSI values in both ME/CFS and control while upright, suggesting improved cerebral autoregulation.


Subject(s)
Blood Pressure , Cerebrovascular Circulation , Orthostatic Intolerance , Phenylephrine , Humans , Cerebrovascular Circulation/drug effects , Phenylephrine/pharmacology , Female , Male , Orthostatic Intolerance/physiopathology , Adult , Young Adult , Blood Flow Velocity/drug effects , Blood Pressure/drug effects , Fatigue Syndrome, Chronic/physiopathology , Fatigue Syndrome, Chronic/drug therapy , Tilt-Table Test , Cognition/drug effects , Homeostasis , Case-Control Studies , Heart Rate/drug effects , Arterial Pressure/drug effects , Postural Orthostatic Tachycardia Syndrome/physiopathology , Postural Orthostatic Tachycardia Syndrome/drug therapy
19.
J Vasc Interv Radiol ; 35(7): 1043-1048.e3, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38508449

ABSTRACT

PURPOSE: Treatment of hypovascular tumors, such as pancreatic adenocarcinoma, is challenging owing to inefficient drug delivery. This report examines the potential mechanism of localized drug delivery via transarterial microperfusion (TAMP) using a proprietary adjustable double-balloon occlusion catheter in a porcine model. MATERIALS AND METHODS: Adult Yorkshire swine (N = 21) were used in the Institutional Animal Care & Use Committee-approved protocols. The RC-120 catheter (RenovoRx, Los Altos, California) was positioned into visceral, femoral, and pulmonary arteries with infusion of methylene blue dye, gemcitabine, or gold nanoparticles. Transmural delivery was compared under double-balloon occlusion with and without side-branch exclusion, single-balloon occlusion, and intravenous delivery. Intra-arterial pressure and vascular histologic changes were assessed. RESULTS: Infusion with double-balloon occlusion and side-branch exclusion provided increased intra-arterial pressure in the isolated segment and enhanced perivascular infusate penetration with minimal vascular injury. Infusates were predominantly found in the vasa vasorum by electron microscopy. CONCLUSIONS: TAMP enhanced transmural passage mediated by localized increase in arterial pressure via vasa vasorum.


Subject(s)
Vasa Vasorum , Animals , Vasa Vasorum/pathology , Vasa Vasorum/drug effects , Balloon Occlusion , Gemcitabine , Infusions, Intra-Arterial , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Models, Animal , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Methylene Blue/administration & dosage , Swine , Metal Nanoparticles , Equipment Design , Arterial Pressure/drug effects , Sus scrofa , Vascular Access Devices
20.
J Cardiovasc Pharmacol ; 83(6): 612-620, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38547510

ABSTRACT

ABSTRACT: Pulmonary arterial hypertension (PAH) is characterized by increased pulmonary vascular resistance (PVR), imposing overload on the right ventricle (RV) and imbalance of the redox state. Our study investigated the influence of treatment with sulforaphane (SFN), found in cruciferous vegetables, on RV remodeling and redox homeostasis in monocrotaline (MCT)-induced PAH. Male Wistar rats were separated into 4 groups: control (CTR); CTR + SFN; MCT; and MCT + SFN. PAH induction was implemented by a single dose of MCT (60 mg/kg intraperitoneally). Treatment with SFN (2.5 mg/kg/day intraperitoneally) started on the seventh day after the MCT injection and persisted for 2 weeks. After 21 days of PAH induction, echocardiographic, hemodynamic, and oxidative stress evaluation was performed. The MCT group showed an increase in RV hypertrophy, RV systolic area, RV systolic, mean pulmonary artery pressure, and PVR and exhibited a decrease in the RV outflow tract acceleration time/ejection time ratio, RV fractional shortening, and tricuspid annular plane systolic excursion compared to CTR ( P < 0.05). SFN-treated PAH attenuated detrimental changes in tricuspid annular plane systolic excursion, mean pulmonary artery pressure, and PVR parameters. Catalase levels and the glutathione/Glutathione disulfide (GSSG) ratio were diminished in the MCT group compared to CTR ( P < 0.05). SFN increased catalase levels and normalized the glutathione/GSSG ratio to control levels ( P < 0.05). Data express the benefit of SFN treatment on the cardiac function of rats with PAH associated with the cellular redox state.


Subject(s)
Disease Models, Animal , Isothiocyanates , Monocrotaline , Oxidation-Reduction , Oxidative Stress , Rats, Wistar , Sulfoxides , Ventricular Function, Right , Animals , Sulfoxides/pharmacology , Isothiocyanates/pharmacology , Male , Ventricular Function, Right/drug effects , Oxidative Stress/drug effects , Antioxidants/pharmacology , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/drug therapy , Homeostasis/drug effects , Ventricular Remodeling/drug effects , Myocardial Contraction/drug effects , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/chemically induced , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Artery/metabolism , Rats , Arterial Pressure/drug effects , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/metabolism , Ventricular Dysfunction, Right/physiopathology , Ventricular Dysfunction, Right/drug therapy , Ventricular Dysfunction, Right/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL