Acessibilidade / Reportar erro

Effects of dexmedetomidine on hemodynamic, oxygenation, microcirculation, and inflammatory markers in a porcine model of sepsis

ABSTRACT

Purpose:

To determine whether dexmedetomidine aggravates hemodynamic, metabolic variables, inflammatory markers, and microcirculation in experimental septic shock.

Methods:

Twenty-four pigs randomized into: Sham group (n = 8), received saline; Shock group (n = 8), received an intravenous infusion of Escherichia coli O55 (3 × 109 cells/mL, 0.75 mL/kg, 1 hour); Dex-Shock group (n = 8), received bacteria and intravenous dexmedetomidine (bolus 0.5 mcg/kg followed by 0.7 mcg/kg/h). Fluid therapy and/ornorepinephrine were administered to maintain a mean arterial pressure > 65 mmHg. Hemodynamic, metabolic, oxygenation, inflammatory markers, and microcirculation were assessed at baseline, at the end of bacterial infusion, and after 60, 120, 180, and 240 minutes.

Results:

Compared to Shock group, Dex-Shock group presented a significantly increased oxygen extraction ratio at T180 (23.1 ± 9.7 vs. 32.5 ± 9.2%, P = 0.0220), decreased central venous pressure at T120 (11.6 ± 1 vs. 9.61 ± 1.2 mmHg, P = 0.0214), mixed-venous oxygen saturation at T180 (72.9 ± 9.6 vs. 63.5 ± 9.2%, P = 0.026), and increased plasma lactate (3.7 ± 0.5 vs. 5.5 ± 1 mmol/L, P = 0.003). Despite the Dex-Shock group having a better sublingual vessel density at T240 (12.5 ± 0.4 vs. 14.4 ± 0.3 mL/m2; P = 0.0003), sublingual blood flow was not different from that in the Shock group (2.4 ± 0.2 vs. 2.4 ± 0.1 mL/kg, P = 0.4418).

Conclusions:

Dexmedetomidine did not worsen the hemodynamic, metabolic, inflammatory, or sublingual blood flow disorders resulting from septic shock. Despite inducing a better sublingual vessel density, dexmedetomidine initially and transitorily increased the mismatch between oxygen supply and demand.

Key words
Sepsis; Dexmedetomidine; Swine

Introduction

Sepsis is frequently observed in critically ill patients and a common cause of mortality. It involves a massive release of inflammatory mediators in response to an injury caused by pathogenic microorganisms in different organs, and it is clinically characterized by arterial hypotension, pulmonary hypertension, endothelial injury, and coagulation disorders. Persistent tissue hypoxia results from microcirculatory impairment and can be followed by the development of ischaemic reperfusion injury and multiple organ failure11 De Backer D, Ricottilli F, Ospina-Tascón GA. Septic shock: a microcirculation disease. Curr Opin Anesthesiol. 2021;34(2):85-91. https://doi.org/10.1097/ACO.0000000000000957
https://doi.org/10.1097/ACO.000000000000...
,22 Sakr Y, Dubois MJ, De Backer D, Creteur J, Vincent JL. Persistent microcirculatory alterations are associated with organ failure and death in patients with septic shock. Crit Care Med. 2004;32(9):1825-1831. https://doi.org/10.1097/01.ccm.0000138558.16257.3f
https://doi.org/10.1097/01.ccm.000013855...
.

Patients in the intensive care unit (ICU) are often uncomfortable because of anxiety, pain, and mechanical ventilation. This discomfort can be treated with analgesics and sedatives, which also facilitate nursing care. Among the different classes of agents, dexmedetomidine is an α2-adrenoceptor agonist with a high affinity for α2-receptors (α2:α1 ratio of 1,620:1), a high potency, and fewer side effects are related to the activation of α1-receptors33 Szumita PM, Baroletti SA, Anger KE, Wechsler ME. Sedation and analgesia in the intensive care unit: evaluating the role of dexmedetomidine. Am J Health Syst Pharm. 2007;64(1):37-44. https://doi.org/10.2146/ajhp050508
https://doi.org/10.2146/ajhp050508...
. The mechanism of action is characterized by the activation of both pre- and postsynaptic α2-adrenoreceptors. Presynaptic activation inhibits the release of norepinephrine and, consequently, modulates pain signalling pathways. In the central nervous system, postsynaptic activation inhibits sympathetic tone, decreasing the heart rate, and blood pressure. The sympatholytic effect reduces the stress response and avoids changes in hemodynamic patterns caused by an increased release of endogenous catecholamines. These combined mechanisms inhibit neuronal firing, produce sedation and analgesia, and decrease nausea, salivation, secretion, and intestinal motility44 Blaudszun G, Lysakowski C, Elia N, Tramèr MR. Effect of perioperative systemic alpha2 agonists on postoperative morphine consumption and pain intensity: systematic review and meta-analysis of randomized controlled trials. Anesthesiology. 2012;116:1312-1322. https://doi.org/10.1097/aln.0b013e31825681cb
https://doi.org/10.1097/aln.0b013e318256...
. The onset of action of dexmedetomidine is observed approximately 15 minutes after the beginning of the infusion, reaching the maximum effect after 1 hour33 Szumita PM, Baroletti SA, Anger KE, Wechsler ME. Sedation and analgesia in the intensive care unit: evaluating the role of dexmedetomidine. Am J Health Syst Pharm. 2007;64(1):37-44. https://doi.org/10.2146/ajhp050508
https://doi.org/10.2146/ajhp050508...
. The synergistic effect with other analgesics reduces the requirement for opioids during surgery and in the postoperative period, decreasing the incidence of respiratory depression caused by opioids33 Szumita PM, Baroletti SA, Anger KE, Wechsler ME. Sedation and analgesia in the intensive care unit: evaluating the role of dexmedetomidine. Am J Health Syst Pharm. 2007;64(1):37-44. https://doi.org/10.2146/ajhp050508
https://doi.org/10.2146/ajhp050508...
,44 Blaudszun G, Lysakowski C, Elia N, Tramèr MR. Effect of perioperative systemic alpha2 agonists on postoperative morphine consumption and pain intensity: systematic review and meta-analysis of randomized controlled trials. Anesthesiology. 2012;116:1312-1322. https://doi.org/10.1097/aln.0b013e31825681cb
https://doi.org/10.1097/aln.0b013e318256...
. In addition, dexmedetomidine has important effects on the immune response, which mainly result from the central sympatholytic effects of dexmedetomidine and its binding to alpha-2 adrenoceptors in macrophages55 Taniguchi T, Kidani Y, Kanakura H, Takemoto Y, Yamamoto K. Effects of dexmedetomidine on mortality rate and inflammatory responses to endotoxin-induced shock in rats. Crit Care Med. 2004;32(6):1322-1326. https://doi.org/10.1097/01.ccm.0000128579.84228.2a
https://doi.org/10.1097/01.ccm.000012857...
,66 Lee JM, Han HJ, Choi WK, Yoo S, Baek S, Lee J. Immunomodulatory effects of intraoperative dexmedetomidine on T helper 1, T helper 2, T helper 17 and regulatory T cells cytokine levels and their balance: a prospective, randomised, double-blind, dose-response clinical study. BMC Anesthesiol. 2018;18:164. https://doi.org/10.1186/s12871-018-0625-2
https://doi.org/10.1186/s12871-018-0625-...
.

Despite the potential benefits of dexmedetomidine for critically ill patients, the drug can also be associated with adverse effects, including initial arterial hypertension followed by hypotension, bradycardia, atrial fibrillation, nausea, and hypoxia. At higher doses, first- and second-degree atrioventricular blockages can be observed33 Szumita PM, Baroletti SA, Anger KE, Wechsler ME. Sedation and analgesia in the intensive care unit: evaluating the role of dexmedetomidine. Am J Health Syst Pharm. 2007;64(1):37-44. https://doi.org/10.2146/ajhp050508
https://doi.org/10.2146/ajhp050508...
,77 Ebert TJ, Hall JE, Barney JA, Uhrich TD, Colinco MD. The effects of increasing plasma concentrations of dexmedetomidine in humans. Anesthesiology. 2000;93:382-394. https://doi.org/10.1097/00000542-200008000-00016
https://doi.org/10.1097/00000542-2000080...
.

Dexmedetomidine facilitates the clinical care of critically ill patients by improving their comfort and preventing delirium88 Su X, Meng ZT, Wu XH, Cui F, Li HL, Wang DX, Zhu X, Zhu SN, Maze M, Ma D. Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: a randomised, double-blind, placebo-controlled trial. Lancet. 2016;388(10054):1893-1902. https://doi.org/10.1016/S0140-6736(16)30580-3
https://doi.org/10.1016/S0140-6736(16)30...
,99 Skrobik Y, Duprey MS, Hill NS, Devlin JW. Low-dose nocturnal dexmedetomidine prevents ICU delirium. A randomized, placebo-controlled trial. Am J Respir Crit Care Med. 2018;197(9):1147-1156. https://doi.org/10.1164/rccm.201710-1995OC
https://doi.org/10.1164/rccm.201710-1995...
. Given the dexmedetomidine-induced cardiovascular and respiratory depression, the benefit/disadvantage ratio for patients with septic shock is unknown. Therefore, the aim of this study was to assess whether dexmedetomidine worsens hemodynamic, oxygenation, metabolic, inflammatory, and microcirculatory responses in a model of septic shock. We hypothesised that dexmedetomidine would not further deteriorate sepsis disorders related disorders or would even improve microcirculatory conditions.

Methods

This prospective randomized experimental study was approved by the Ethics Committee for research projects at our institution (#1,420/2008). All animals received human care in compliance with the Principles of Laboratory Animal Care formulated by the National Society for Medical Research and the US National Academy of Sciences Guide for the Care and Use of Laboratory Animals.

Animal preparation

Twenty-four Landrace and Large White crossbred female pigs weighing 24.1 ± 2.4 kg were used in the study. The animals were fasted for 12 hours with free access to water before the experiments. Animals were premedicated with midazolam (0.25 mg/kg) and ketamine (5 mg/kg) intramuscularly. Anaesthesia was induced with propofol (5 mg/kg) administered intravenously (IV) and, after endotracheal intubation, maintained with isoflurane (1.4% end-tidal concentration) vaporized in 40% oxygen. Pancuronium was administrated (bolus of 0.1 mg/kg followed by infusion of 0.02 mg/kg/min), and mechanical ventilation (Primus; Dräger, Lübeck, Germany) was performed using the volume-controlled ventilation mode with a tidal volume of 8 mL/kg, a positive end-expiratory pressure (PEEP) of 5 cmH2O and the respiratory rate adjusted to maintain an end-tidal carbon dioxide (ETCO2) between 35-45 mmHg. Local anaesthesia was performed by administering 3 mL of 2% lidocaine at each incision site. Lactated Ringer’s solution was administered at 5 mL/kg/h during preparation and at 10 mL/kg/hduring the experimentation period. Body temperature was maintained between 37-38 °C by using a heated mat (Medi-therm II; Gaymar Industries, Orchard Park, NY, United States of America).

Experimental protocol

Bacterial preparation

A strain of Escherichia coli (EPEC, O55) from VPS-FMVZ-USP was activated in trypticase soy broth (TSB) for 24 hours, spread on trypticase soy agar (TSA), and incubated for 24 hours at 37 °C. After bacterial growth, aliquots were suspended and diluted in saline to obtain a solution of 3 × 1099 Skrobik Y, Duprey MS, Hill NS, Devlin JW. Low-dose nocturnal dexmedetomidine prevents ICU delirium. A randomized, placebo-controlled trial. Am J Respir Crit Care Med. 2018;197(9):1147-1156. https://doi.org/10.1164/rccm.201710-1995OC
https://doi.org/10.1164/rccm.201710-1995...
cells/mL, which corresponded to 0.6 × 101010 Garrido AG, Poli de Figueiredo LF, Cruz RJ Jr., Silva E, Rocha E Silva M. Short-lasting systemic and regional benefits of early crystalloid infusion after intravenous inoculation of dogs with live Escherichia coli. Braz J Med Biol Res. 2005;38(6):873-884. https://doi.org/10.1590/s0100-879x2005000600009
https://doi.org/10.1590/s0100-879x200500...
ufc/mL/live E. coli. The target concentration of bacteria was measured via spectrophotometry, with a final absorbance between 0.990 and 0.9601010 Garrido AG, Poli de Figueiredo LF, Cruz RJ Jr., Silva E, Rocha E Silva M. Short-lasting systemic and regional benefits of early crystalloid infusion after intravenous inoculation of dogs with live Escherichia coli. Braz J Med Biol Res. 2005;38(6):873-884. https://doi.org/10.1590/s0100-879x2005000600009
https://doi.org/10.1590/s0100-879x200500...
. The bacteria solution was stored at 4 °C for 12 to 36 hours prior to IV administration to the animals.

Experimental design

Following surgical preparation, baseline data were obtained, and animals were randomly allocated into one of the following three groups:

  • a Shock group (n = 8) consisting of animals that received a 0.75-mL/kg infusion of E. coli O55 solution for 60 minutes1111 Rahal L, Garrido AG, Cruz RJ Jr, Silva E, Poli-de-Figueiredo LF. Fluid replacement with hypertonic or isotonic solutions guided by mixed venous oxygen saturation in experimental hypodynamic sepsis. J Trauma. 2009;67(6):1205-1212. https://doi.org/10.1097/TA.0b013e31818b2567
    https://doi.org/10.1097/TA.0b013e31818b2...
    ;

  • a Dex-Shock group (n = 8), that simultaneously received infusions of bacteria and dexmedetomidine (bolus of 0.5 μg/kgin 10 minutes, followed by a constant rate infusion of 0.7 μg/kg/h until the end of the experiment);

  • a Sham group (n = 8), that did not receive the bacteria or dexmedetomidine infusion.

The Sham and Shock groups received saline solution at an infusion rate equivalent to that of dexmedetomidine. Randomization was previously performed, and the group allocation was blindly placed in numbered manila envelopes, which were opened in a consecutive manner immediately before baseline measurements were registered.

After sepsis induction, the animals were monitored and treated from T0 to T240. A bolus of 20 mL/kg lactated Ringer’s solution was infused within 20 minutes if they had arterial hypotension (mean arterial pressure – MAP < 65 mmHg), central venous pressure (CVP) ≤ 12 mmHg, mixed-venous oxygen saturation (SvO2) < 65% and urine output < 0.5 mL/kg/h. If these alterations were present with a CVP > 12 mmHg, the animals received a norepinephrine infusion (starting rate of 0.1 μg/kg/min, with the dose increasing by 0.05 μg/kg/min every 5 minutes, for up to 2 μg/kg/min) until hemodynamic stabilization was achieved1212 Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R, Reinhart K, Angus DC, Brun-Buisson C, Beale R, Calandra T, Dhainaut JF, Gerlach H, Harvey M, Marini JJ, Marshall J, Ranieri M, Ramsay G, Sevransky J, Thompson BT, Townsend S, Vender JS, Zimmerman JL, Vincent JL. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock. Crit Care Med. 2008;36(1):296-327. https://doi.org/10.1097/01.CCM.0000298158.12101.41
https://doi.org/10.1097/01.CCM.000029815...
. The volume of additional fluids, norepinephrine requirements, and urine output were recorded (Fig. 1).

Figure 1
Experimental design.

Measurements

Hemodynamics and blood gas analysis

Both the femoral artery and vein were catheterized for arterial pressure monitoring, blood sampling, and fluid administration. A 7.5-F pulmonary artery catheter (Swan-Ganz; Edwards Lifesciences, Irvine, CA, United States of America) was surgically introduced into the right internal jugular vein and advanced under continuous pressure recording into wedge position. Cardiac output was determined by the thermodilution method (Vigilance monitor; Edwards Lifesciences). The cardiac index (CI) was calculated to normalize the data for body surface area in square meters by using a conversion factor appropriate for pigs (Eq. 1):

k   ×   BW 2 / 3 (1)

In which: k = 0.09; BW = body weight in kg1313 Holt JP, Rhode EA, Kines H. Ventricular volumes and body weight in mammals. Am J Physiol. 1968;215:704-715. HTTPS://DOI.ORG/10.1152/ajplegacy.1968.215.3.704.
https://doi.org/10.1152/ajplegacy.1968.2...
.

The heart rate (HR), MAP, CVP, mean pulmonary artery pressure (MPAP), and pulmonary artery occlusion pressure (PAOP) were continuously monitored with a multiparametric monitor (IntelliVue MP50, Philips Healthcare, Best, Netherlands). The systemic vascular resistance index (SVRI), pulmonary vascular resistance index (PVRI), stroke volume index (SVI), systemic oxygen delivery index (DO2I), systemic oxygen consumption index (VO2I), and systemic oxygen extraction ratio (O2ER) were calculated utilizing standard formulae. Arterial and mixed venous blood samples were collected simultaneously at each time point and immediately analysed (ABL 555; Radiometer, Copenhagen, Denmark) for blood gas analyses, including measurement of haemoglobin (Hb), lactate, and potassium (K+). Blood glucose was assessed with a portable device (Accu-Check Advantage II; Roche, Mannheim, Germany). A 5%-glucose solution was used when necessary to maintain blood glucose > 40 mg/dL. The volume of 5% glucose solution administered, if any, was recorded.

Sublingual microcirculation assessment and jejunal tonometry

In-vivo microscopy of the sublingual mucosa was performed using the orthogonal polarization spectral (OPS) technique (MicroScan®, MicroVision Medical Inc.). Five sequences of 20 seconds each were recorded at each time point using a digital image conversion device. The sequences were analysed for vessel density and blood flow using AVA 3.0 software.

A tonometer tube with a silicone rubber balloon (catheter TRIP NGS, Tonometrics, Worcester) was inserted into the jejunum via a laparotomy to measure intestinal mucosal carbon dioxide (PrCO2) using air-automated tonometry (Tonocap, Datex, Helsinki). Arterial pH and PaCO2 values measured at the same time were used to calculate the intestinal pH (pHi) and intestinal mucosal-to-arterial carbon dioxide pressure difference (Pr-aCO2).

Biological markers of inflammation

The blood samples were centrifuged at 2,000 rpm for 10 minutes at 4 °C. The plasma was stored at -80 °C until analysis. The plasma concentrations of tumour necrosis factor alpha (TNF-α) and interleukins 1β (IL-1β), 6 (IL-6), and 10 (IL-10) were measured using enzyme linked immunosorbent (ELISA) assays according to the manufacturer’s instructions (DuoSet®, ELISA Development System, R&D Systems, Minneapolis, United States of America). The plasma levels of each cytokine were obtained through optical density measurements, and the absorbance was converted to pg/mL using a nonlinear regression curve and a standard curve.

Cortisol was measured using commercial immunoassay kits (Autodelfia Cortisol Kit, Wallac, Finland).

Data acquisition

The hemodynamic, jejunal tonometry, and blood gas data were measured prior to the bacterial infusion (baseline); immediately after infusion (T0); and 60 (T60), 120 (T120), 180 (T180), and 240 minutes (T240) later. Blood samples for cytokine measurements were obtained at baseline, T0, T60, and T240. The sublingual OPS images were recorded at baseline, T0, and T240 (Fig. 1). At the end of the experiment, isoflurane was increased to 5%, and the animals were euthanized via administration of an intravenous injection of potassium chloride.

Statistical analysis

The sample size for paired data was calculated using power analysis. A minimum of eight pigs per group was required to have a 95% chance (with 5% risk) to detect a difference of 3.5 mmol/L in blood lactate between groups, considering a standard deviation of 2 mmol/L. All data were assessed for normality using a D’Agostino-Pearson’s test. Body weight, urine output, fluid volume, and norepinephrine consumption were compared between groups using one-way analysis of variance (ANOVA) and Student’s t test. Normally distributed data were analysed within groups and among groups using two-way ANOVA for repeated measures (Sham vs. Shock and Dex-Shock, and then Shock vs. Dex-Shock) with a post hoc Tukey’s test when appropriate. Non-normally distributed data were compared within groups using a Friedman’s test with a post hoc Dunn’s test, and the analysis between groups was performed using a Kruskal-Wallis and post hoc Dunn’s test. Statistical significance was defined as p < 0.05. All tests were performed using a statistical software (Prism 6 for Windows, GraphPad). The results are presented as the mean ± standard deviation or median (interquartile range).

Results

Body weight was not significantly different between groups (Sham: 24.3 ± 2.6 kg; Shock: 24.7 ± 3 kg; Dex-Shock: 23.4 ± 1.3 kg, p = 0.5558).

Septic shock-induced disorders

Fluid loading, norepinephrine administration, and urinary output

The Sham group did not require additional fluid therapy or norepinephrine infusion throughout the study (Fig. 2). The fluid requirement (100 ± 22 mL/kg), and norepinephrine consumption (64.15 ± 93.4 μg/kg) were significantly higher in the Shock group than in the Sham group (50 ± 0 mL/kg and 0 μg/kg, respectively). There was no significant difference in urine output between groups (p = 0.0757).

Figure 2
Fluid loading, total dose of norepinephrine, and urinary output in anaesthetised pigs (Sham group), septic shock animals (Shock group), and septic shock animals receiving dexmedetomidine (Dex-Shock group).

Hemodynamic and microcirculation disorders

The Shock group had a significant increase (at T0, T60, T120, T180, and T240) in HR, MPAP, CVP, and PVRI, and a significant decrease in SVI (at T0, T120, T180, and T240) compared with baseline (Fig. 3). The SVRI was significantly decreased only at T60 in the Shock group (p = 0.0424). The CI increased significantly only at T60 in the Shock group (p = 0.0002). No significant hemodynamic changes were observed in Sham animals.

Figure 3
Hemodynamic changes in anesthetized pigs (Sham group), septic shock animals (Shock group), and septic shock animals administered dexmedetomidine (Dex-Shock group) before bacterial infusion (baseline); at the end of bacterial infusion (T0); and 60 (T60), 120 (T120), 180 (T180), and 240 minutes (T240) after bacterial infusion.

Sublingual blood flow was significantly reduced by septic shock (p = 0.008), whereas vessel density was not altered (p = 0.2851) (Fig. 4). The intestinal regional pH decreased significantly in the Shock group (p < 0.0001). Sham animals did not show any significant microcirculatory changes.

Figure 4
Sublingual microcirculation and jejunal tonometry in anesthetized pigs (Sham group), septic shock animals (Shock group), and septic shock animals administered a dexmedetomidine infusion (Dex-Shock group). (a) An illustrative example for sublingual vessels density. (b) The median and 25-75 percentile values for sublingual blood flow and vessel density before bacterial infusion, at the end of bacterial infusion (T0), and 240 minutes (T240) after bacterial infusion. (c) The meanvalues for intestinal pH (pHi) and intestinal mucosal-to-arterial carbon dioxide pressure difference (Pr-aCO2).

Systemic oxygenation, blood gas and electrolytes

Septic shock resulted in a significant decrease in the arterial pH (from T0 to T240), the PaO2/FiO2 ratio (from T0 to T240), and plasma bicarbonate (from T60 to T240), and a significant increase in the plasma lactate (from T60 to T240), K+ (T240), and haematocrit (at T0, T120, T180 and T240) compared with baseline (Table 1 and Fig. 5). These variables were also significantly different compared with those in Sham animals.

Table 1
Blood gas and electrolytes.
Figure 5
Changes in oxygen consumption (VO2I), the oxygen extraction ratio (O2ER), mixed venous oxygen saturation (SvO2), and plasma lactate in anaesthetised pigs (Sham group), septic shock animals (Shock group), and septicshock plus dexmedetomidine infusion animals (Dex-Shock group) at baseline; at the end of bacterial infusion(T0); and after 60 (T60), 120 (T120), 180 (T180), and 240 minutes (T240) after bacterial infusion.

Blood glucose was not significantly changed in the Shock group compared with baseline. In the Sham group, blood glucose showed a slight increase at T60 compared with baseline. No significant differences in blood glucose were observed between Sham group and both Shock groups.

Inflammatory markers

Animals in the Shock group exhibited a significant increase in the plasma levels of TNF-α (T0, T60, and T240), IL-1β (T240), IL-6 (T60 and T240), IL-10 (T0), and cortisol (T0 and T240) compared with baseline (Table 2). Sham animals showed no significant changes in inflammatory markers.

Table 2
Plasma cytokines and cortisol.

Impact of dexmedetomidine on septic shock-induced disorders

Fluid loading, norepinephrine administration, and urinary output

Fluid loading (p = 0.5848), norepinephrine requirements (p = 0.8438), and urine output (p = 0.1916) were not significantly different between the Shock and Dex-Shock groups (Fig. 2).

Hemodynamic and microcirculatory disorders

The infusion of dexmedetomidine did not modify septic shock-induced cardiorespiratory disorders or sublingual blood flow (Table 1 and Fig. 3). However, the blood vessel density was significantly higher at T240 in the Dex-Shock group than in the untreated Shock group (p = 0.0126; Fig. 4).

Systemic oxygenation, blood gas, and electrolytes

Septic shock resulted in a decrease in the arterial pH, the PaO2/FiO2 ratio, plasma bicarbonate, and pHi, and an increase in O2ER, the haematocrit, and K+ (Table 1 and Fig. 5). These alterations were not significantly modified by the intravenous infusion of dexmedetomidine.

Inflammatory markers

Dexmedetomidine did not modify the septic shock-induced increase in TNF-α, IL-1β, IL-6, IL-10, or cortisol (Table 2).

Discussion

In this study, performed in anesthetized pigs injected with live E. coli and monitored over 4 hours, dexmedetomidine did not impact norepinephrine requirements; did not mitigate or worsen septic shock-induced hemodynamic disorders; promoted a slight increase in impact sublingual vessel density, and induced an initial and transitory increase in oxygen consumption and a late decrease in mixed venous O2 saturation associated with a slight but significant increase in lactate.

The intravenous administration of live E. coli caused septic shock characterized by an immediate decrease in the MAP requiring norepinephrine administration, a severe reduction in urinary output, and an initial hyperdynamic state followed by a progressive and continuous decrease in the cardiac index. Dexmedetomidine did not modify norepinephrine requirement, but preserved urinary output. This result confirms previous experimental and clinical studies reporting that dexmedetomidine can offer a protective effect against septic1414 Qiu R, Yao W, Ji H, Yuan D, Gao X, Sha W, Wang F, Huang P, Hei Z. Dexmedetomidine restores septic renal function via promoting inflammation resolution in a rat sepsis model. Life Sci. 2018;204:1-8. https://doi.org/10.1016/j.lfs.2018.05.001
https://doi.org/10.1016/j.lfs.2018.05.00...
and postoperative acute kidney injury1515 Cho JS, Shim JK, Soh S, Kim MK, Kwak YL. Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery. Kidney Int. 2016;89(3):693-700. https://doi.org/10.1038/ki.2015.306
https://doi.org/10.1038/ki.2015.306...
,1616 Liu Y, Sheng B, Wang S, Lu F, Zhen J, Chen W. Dexmedetomidine prevents acute kidney injury after adult cardiac surgery: a meta-analysis of randomized controlled trials. BMC Anesthesiol. 2018;18:7. https://doi.org/10.1186/s12871-018-0472-1
https://doi.org/10.1186/s12871-018-0472-...
by exerting an anti-inflammatory effect and ischemia/reperfusion attenuation. Dexmedetomidine also inhibits the release of vasopressin and insulin, increasing urinary output and blood glucose1717 Sinclair MD. A review of the physiological effects of alpha2-agonists related to the clinical use of medetomidine in small animal practice. Can Vet J. 2003;44(11):885-897.,1818 Villela NR, Nascimento Júnior P, Carvalho LR, Teixeira A. Effects of dexmedetomidine on renal system and on vasopressin plasma levels. Experimental study in dogs. Rev Bras Anestesiol. 2005;55(4):429-440. https://doi.org/10.1590/s0034-70942005000400007
https://doi.org/10.1590/s0034-7094200500...
. Because α2-agonists improve the pressor response to norepinephrine1919 Geloen A, Chapelier K, Cividjian A, Dantony E, Rabilloud M, May CN, Quintin L. Clonidine and dexmedetomidine increase the pressor response to norepinephrine in experimental sepsis: a pilot study. Crit Care Med. 2013;41(12):e431-8. https://doi.org/10.1097/CCM.0b013e3182986248
https://doi.org/10.1097/CCM.0b013e318298...
, reduced vasoactive drug requirements during septic shock have been reported in patients sedated by dexmedetomidine2020 Morelli A, Sanfilippo F, Arnemann P, Hessler M, Kampmeier TG, D’Egidio A, Orecchioni A, Santonocito C, Frati G, Greco E, Westphal M, Rehberg SW, Ertmer C. The effect of propofol and dexmedetomidine sedation on norepinephrine requirements in septic shock patients: a crossover trial. Crit Care Med. 2019;47(2):e89-e95. https://doi.org/10.1097/CCM.0000000000003520
https://doi.org/10.1097/CCM.000000000000...
. However, this benefit is inconsistently observed2121 Nelson KM, Patel GP, Hammond DA. Effects from continuous infusions of dexmedetomidine and propofol on hemodynamic stability in critically Ill adult patients with septic shock. J Intensive Care Med. 2020;35(9):875-880. https://doi.org/10.1177/0885066618802269
https://doi.org/10.1177/0885066618802269...
and was not documented in the present study. Dexmedetomidine reversed the septic shock-induced increase in CVP from the second hour following bacterial injection, and did not modify the other septic shock-induced hemodynamic disorders. The alterations in sublingual and intestinal microcirculation induced by septic shock11 De Backer D, Ricottilli F, Ospina-Tascón GA. Septic shock: a microcirculation disease. Curr Opin Anesthesiol. 2021;34(2):85-91. https://doi.org/10.1097/ACO.0000000000000957
https://doi.org/10.1097/ACO.000000000000...
were not worsened by dexmedetomidine. Additionally, in the present study, dexmedetomidine did not prevent the decrease in sublingual blood flow, but significantly increased vessel density. This finding is in accordance with a previous study showing that dexmedetomidine attenuates the microcirculatory derangements associated with experimental sepsis2222 Miranda ML, Balarini MM, Bouskela E. Dexmedetomidine attenuates the microcirculatory derangements evoked by experimental sepsis. Anesthesiology. 2015;122:619-630. https://doi.org/10.1097/ALN.0000000000000491
https://doi.org/10.1097/ALN.000000000000...
. The mechanisms are not fully understood, but leukocyte rolling, and adhesion may be involved2222 Miranda ML, Balarini MM, Bouskela E. Dexmedetomidine attenuates the microcirculatory derangements evoked by experimental sepsis. Anesthesiology. 2015;122:619-630. https://doi.org/10.1097/ALN.0000000000000491
https://doi.org/10.1097/ALN.000000000000...
.

Despite the lack of improvement in sublingual and intestinal blood flows, dexmedetomidine induced an initial tissue O2 impairment reflected by a significant increase in O2ER and a late increase in lactate associated with a significant decrease in SvO2. This result is more relevant because dexmedetomidine increases lactate clearance in patients with septic shock2323 Miyamoto K, Nakashima T, Shima N, Kato S, Ueda K, Kawazoe Y, Ohta Y, Morimoto T, Yamamura H; DESIRE Trial Investigators. Effect of dexmedetomidine on lactate clearance in patients with septic shock: a subanalysis of a multicenter randomized controlled trial. Shock. 2018;50(2):162-166. https://doi.org/10.1097/SHK.0000000000001055
https://doi.org/10.1097/SHK.000000000000...
. SvO2 has been shown to be a surrogate for the cardiac index, a target for hemodynamic therapy2424 Gattinoni L, Brazzi L, Pelosi P, Latini R, Tognoni G, Pesenti A, Fumagalli R. A trial of goal-oriented hemodynamic therapy in critically ill patients. SvO2 Collaborative Group. N Engl J Med. 1995;333:1025-1032. https://doi.org/10.1056/NEJM199510193331601
https://doi.org/10.1056/NEJM199510193331...
. Accordingly, in the present study, the decrease in SvO2 in the Dex-Shock group reflected the decrease in the cardiac index, which was higher in the middle of the experiment, but decreased at the end of it. Although the current dose of dexmedetomidine does not modify the cardiac index in healthy animals2525 Pascoe PJ. The cardiopulmonary effects of dexmedetomidine infusions in dogs during isoflurane anesthesia. Vet Anaesth Analg. 2015;42(4):360-368. https://doi.org/10.1111/vaa.12220
https://doi.org/10.1111/vaa.12220...
, it might have an impact in the presence of sepsis1919 Geloen A, Chapelier K, Cividjian A, Dantony E, Rabilloud M, May CN, Quintin L. Clonidine and dexmedetomidine increase the pressor response to norepinephrine in experimental sepsis: a pilot study. Crit Care Med. 2013;41(12):e431-8. https://doi.org/10.1097/CCM.0b013e3182986248
https://doi.org/10.1097/CCM.0b013e318298...
. In addition, the haemoconcentration caused by fluid extravasation from the microcirculation may have contributed to the development of a compensatory increase in oxygen extraction, which consequently led to the SvO2 and cardiac index decrease. The deterioration of systemic oxygenation was accompanied by changes in arterial lactate and pH, which were consistent with metabolic lactic acidosis. As attested by the development of splanchnic acidosis, tissue oxygenation was impaired in all septic animals, confirming a previous study1010 Garrido AG, Poli de Figueiredo LF, Cruz RJ Jr., Silva E, Rocha E Silva M. Short-lasting systemic and regional benefits of early crystalloid infusion after intravenous inoculation of dogs with live Escherichia coli. Braz J Med Biol Res. 2005;38(6):873-884. https://doi.org/10.1590/s0100-879x2005000600009
https://doi.org/10.1590/s0100-879x200500...
. However, dexmedetomidine did not further affect the intestinal pH, PrCO2, or Pr-aCO2, as previously reported in septic patients2626 Memiş D, Hekimoğlu S, Vatan I, Yandim T, Yüksel M, Süt N. Effects of midazolam and dexmedetomidine on inflammatory responses and gastric intramucosal pH to sepsis, in critically ill patients. Br J Anaesth. 2007;98(4):550-552. https://doi.org/10.1093/bja/aem017
https://doi.org/10.1093/bja/aem017...
. Therefore, our data do not allow us to identify dexmedetomidine-induced tissue O2 impairment.

As previously reported, the intravenous injection of live E. coli induced an increase in the pulmonary artery pressure and pulmonary vascular resistance1010 Garrido AG, Poli de Figueiredo LF, Cruz RJ Jr., Silva E, Rocha E Silva M. Short-lasting systemic and regional benefits of early crystalloid infusion after intravenous inoculation of dogs with live Escherichia coli. Braz J Med Biol Res. 2005;38(6):873-884. https://doi.org/10.1590/s0100-879x2005000600009
https://doi.org/10.1590/s0100-879x200500...
,2626 Memiş D, Hekimoğlu S, Vatan I, Yandim T, Yüksel M, Süt N. Effects of midazolam and dexmedetomidine on inflammatory responses and gastric intramucosal pH to sepsis, in critically ill patients. Br J Anaesth. 2007;98(4):550-552. https://doi.org/10.1093/bja/aem017
https://doi.org/10.1093/bja/aem017...
; a significant decrease in PaO2/FiO2, and a significant increase in PaCO2, in contrast to several experimental studies reporting that dexmedetomidine attenuates endotoxin and ventilator-induced lung injury2727 Chen H, Sun X, Yang X, Hou Y, Yu X, Wang Y, Wu J, Liu D, Wang H, Yu J, Yi W. Dexmedetomidine reduces ventilator-induced lung injury (VILI) by inhibiting Toll-like receptor 4 (TLR4)/nuclear factor (NF)-κB signaling pathway. Bosn J Basic Med Sci. 2018;18(2):162-169. https://doi.org/10.17305/bjbms.2018.2400
https://doi.org/10.17305/bjbms.2018.2400...

28 Yang CL, Chen CH, Tsai PS, Wang TY, Huang CJ. Protective effects of dexmedetomidine-ketamine combination against ventilator-induced lung injury in endotoxemia rats. J Surg Res. 2011;167(2):e273-81. https://doi.org/10.1016/j.jss.2010.02.020
https://doi.org/10.1016/j.jss.2010.02.02...
-2929 Yang CL, Tsai PS, Huang CJ. Effects of dexmedetomidine on regulating pulmonary inflammation in a rat model of ventilator-induced lung injury. Acta Anaesthesiol Taiwan. 2008;46(4):151-159. https://doi.org/10.1016/S1875-4597(09)60002-3
https://doi.org/10.1016/S1875-4597(09)60...
. In our study, dexmedetomidine did not modify any of the respiratory disorders resulting from the intravenous injection of live E. coli.

Intravenous injection of E. coli also induces the release of inflammatory cytokines3030 Calzavacca P, Booth LC, Lankadeva YR, Bailey SR, Burrell LM, Bailey M, Bellomo R, May CN. Effects of clonidine on the cardiovascular, renal, and Inflammatory responses to experimental bacteremia. Shock. 2019;51(3):348-355. https://doi.org/10.1097/SHK.0000000000001134
https://doi.org/10.1097/SHK.000000000000...
,3131 Thorgersen EB, Hellerud BC, Nielsen EW, Barratt-Due A, Fure H, Lindstad JK, Pharo A, Fosse E, Tønnessen TI, Johansen HT, Castellheim A, Mollnes TE. CD14 inhibition efficiently attenuates early inflammatory and hemostatic responses in Escherichia coli sepsis in pigs. FASEB J. 2010;24(3):712-722. https://doi.org/10.1096/fj.09-140798
https://doi.org/10.1096/fj.09-140798...
. In our study, dexmedetomidine did not modify cytokine release, although previous experimental studies reported a dose-dependent decrease in TNF-α and IL-6 in an endotoxin-induced shock model55 Taniguchi T, Kidani Y, Kanakura H, Takemoto Y, Yamamoto K. Effects of dexmedetomidine on mortality rate and inflammatory responses to endotoxin-induced shock in rats. Crit Care Med. 2004;32(6):1322-1326. https://doi.org/10.1097/01.ccm.0000128579.84228.2a
https://doi.org/10.1097/01.ccm.000012857...
,3232 Taniguchi T, Kurita A, Kobayashi K, Yamamoto K, Inaba H. Dose- and time-related effects of dexmedetomidine on mortality and inflammatory responses to endotoxin-induced shock in rats. J Anesth. 2008;22:221-228. https://doi.org/10.1007/s00540-008-0611-9
https://doi.org/10.1007/s00540-008-0611-...
. In vitro, dexmedetomidine failed to influence the cytokine levels and neutrophil function associated with chemotaxis, phagocytosis, or superoxide production after E. coli exposure3333 Nishina K, Akamatsu H, Mikawa K, Shiga M, Maekawa N, Obara H, Niwa Y. The effects of clonidine and dexmedetomidine on human neutrophil functions. Anesth Analg. 1999;88(2):452-458. https://doi.org/10.1097/00000539-199902000-00042
https://doi.org/10.1097/00000539-1999020...
.

However, in a clinical trial, septic patients sedated with dexmedetomidine had lower levels of TNF-α, IL-1β, and IL-6 detected 24 hours after admission to the ICU2626 Memiş D, Hekimoğlu S, Vatan I, Yandim T, Yüksel M, Süt N. Effects of midazolam and dexmedetomidine on inflammatory responses and gastric intramucosal pH to sepsis, in critically ill patients. Br J Anaesth. 2007;98(4):550-552. https://doi.org/10.1093/bja/aem017
https://doi.org/10.1093/bja/aem017...
. Therefore, the lack of a significant reduction in cytokine levels in the present study may be related to the insufficient assessment time after E. coli infusion. In addition, the cytokine response is characterized by large individual variability, and the absence of a significant impact of dexmedetomidine on IL-6 and IL-10 might be related to insufficient power, as previously reported3434 Venn RM, Bryant A, Hall GM, Grounds RM. Effects of dexmedetomidine on adrenocortical function, and the cardiovascular, endocrine and inflammatory responses in post-operative patients needing sedation in the intensive care unit. Br J Anaesth. 2001;86(5):650-656. https://doi.org/10.1093/bja/86.5.650
https://doi.org/10.1093/bja/86.5.650...
.

Conclusions

Dexmedetomidine did not affect the early hemodynamic, metabolic, and inflammatory disorders induced by septic shock. However, a late mismatch between oxygen supply and demand was observed in animals receiving dexmedetomidine, which can also be caused by cardiac output reduction. Finally, dexmedetomidine preserved the sublingual microcirculatory vessel density, but it did not protect against septic shock-induced decrease in sublingual blood flow. Therefore, the results of the present study suggest that dexmedetomidine should be used cautiously in septic shock patients.

Acknowledgments

Not applicable.

  • Research performed at Surgery Department, Faculdade de Medicina Veterinária e Zootecnia, and LIM08-Laboratory of Anesthesiology, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo (SP), Brazil.
  • Funding

    Fundação de Amparo . Pesquisa do Estado de São Paulo.
    Grant nº 08/58875-4
  • Data availability statement

    Data will be available upon request.

References

  • 1
    De Backer D, Ricottilli F, Ospina-Tascón GA. Septic shock: a microcirculation disease. Curr Opin Anesthesiol. 2021;34(2):85-91. https://doi.org/10.1097/ACO.0000000000000957
    » https://doi.org/10.1097/ACO.0000000000000957
  • 2
    Sakr Y, Dubois MJ, De Backer D, Creteur J, Vincent JL. Persistent microcirculatory alterations are associated with organ failure and death in patients with septic shock. Crit Care Med. 2004;32(9):1825-1831. https://doi.org/10.1097/01.ccm.0000138558.16257.3f
    » https://doi.org/10.1097/01.ccm.0000138558.16257.3f
  • 3
    Szumita PM, Baroletti SA, Anger KE, Wechsler ME. Sedation and analgesia in the intensive care unit: evaluating the role of dexmedetomidine. Am J Health Syst Pharm. 2007;64(1):37-44. https://doi.org/10.2146/ajhp050508
    » https://doi.org/10.2146/ajhp050508
  • 4
    Blaudszun G, Lysakowski C, Elia N, Tramèr MR. Effect of perioperative systemic alpha2 agonists on postoperative morphine consumption and pain intensity: systematic review and meta-analysis of randomized controlled trials. Anesthesiology. 2012;116:1312-1322. https://doi.org/10.1097/aln.0b013e31825681cb
    » https://doi.org/10.1097/aln.0b013e31825681cb
  • 5
    Taniguchi T, Kidani Y, Kanakura H, Takemoto Y, Yamamoto K. Effects of dexmedetomidine on mortality rate and inflammatory responses to endotoxin-induced shock in rats. Crit Care Med. 2004;32(6):1322-1326. https://doi.org/10.1097/01.ccm.0000128579.84228.2a
    » https://doi.org/10.1097/01.ccm.0000128579.84228.2a
  • 6
    Lee JM, Han HJ, Choi WK, Yoo S, Baek S, Lee J. Immunomodulatory effects of intraoperative dexmedetomidine on T helper 1, T helper 2, T helper 17 and regulatory T cells cytokine levels and their balance: a prospective, randomised, double-blind, dose-response clinical study. BMC Anesthesiol. 2018;18:164. https://doi.org/10.1186/s12871-018-0625-2
    » https://doi.org/10.1186/s12871-018-0625-2
  • 7
    Ebert TJ, Hall JE, Barney JA, Uhrich TD, Colinco MD. The effects of increasing plasma concentrations of dexmedetomidine in humans. Anesthesiology. 2000;93:382-394. https://doi.org/10.1097/00000542-200008000-00016
    » https://doi.org/10.1097/00000542-200008000-00016
  • 8
    Su X, Meng ZT, Wu XH, Cui F, Li HL, Wang DX, Zhu X, Zhu SN, Maze M, Ma D. Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: a randomised, double-blind, placebo-controlled trial. Lancet. 2016;388(10054):1893-1902. https://doi.org/10.1016/S0140-6736(16)30580-3
    » https://doi.org/10.1016/S0140-6736(16)30580-3
  • 9
    Skrobik Y, Duprey MS, Hill NS, Devlin JW. Low-dose nocturnal dexmedetomidine prevents ICU delirium. A randomized, placebo-controlled trial. Am J Respir Crit Care Med. 2018;197(9):1147-1156. https://doi.org/10.1164/rccm.201710-1995OC
    » https://doi.org/10.1164/rccm.201710-1995OC
  • 10
    Garrido AG, Poli de Figueiredo LF, Cruz RJ Jr., Silva E, Rocha E Silva M. Short-lasting systemic and regional benefits of early crystalloid infusion after intravenous inoculation of dogs with live Escherichia coli. Braz J Med Biol Res. 2005;38(6):873-884. https://doi.org/10.1590/s0100-879x2005000600009
    » https://doi.org/10.1590/s0100-879x2005000600009
  • 11
    Rahal L, Garrido AG, Cruz RJ Jr, Silva E, Poli-de-Figueiredo LF. Fluid replacement with hypertonic or isotonic solutions guided by mixed venous oxygen saturation in experimental hypodynamic sepsis. J Trauma. 2009;67(6):1205-1212. https://doi.org/10.1097/TA.0b013e31818b2567
    » https://doi.org/10.1097/TA.0b013e31818b2567
  • 12
    Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R, Reinhart K, Angus DC, Brun-Buisson C, Beale R, Calandra T, Dhainaut JF, Gerlach H, Harvey M, Marini JJ, Marshall J, Ranieri M, Ramsay G, Sevransky J, Thompson BT, Townsend S, Vender JS, Zimmerman JL, Vincent JL. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock. Crit Care Med. 2008;36(1):296-327. https://doi.org/10.1097/01.CCM.0000298158.12101.41
    » https://doi.org/10.1097/01.CCM.0000298158.12101.41
  • 13
    Holt JP, Rhode EA, Kines H. Ventricular volumes and body weight in mammals. Am J Physiol. 1968;215:704-715. HTTPS://DOI.ORG/10.1152/ajplegacy.1968.215.3.704.
    » https://doi.org/10.1152/ajplegacy.1968.215.3.704
  • 14
    Qiu R, Yao W, Ji H, Yuan D, Gao X, Sha W, Wang F, Huang P, Hei Z. Dexmedetomidine restores septic renal function via promoting inflammation resolution in a rat sepsis model. Life Sci. 2018;204:1-8. https://doi.org/10.1016/j.lfs.2018.05.001
    » https://doi.org/10.1016/j.lfs.2018.05.001
  • 15
    Cho JS, Shim JK, Soh S, Kim MK, Kwak YL. Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery. Kidney Int. 2016;89(3):693-700. https://doi.org/10.1038/ki.2015.306
    » https://doi.org/10.1038/ki.2015.306
  • 16
    Liu Y, Sheng B, Wang S, Lu F, Zhen J, Chen W. Dexmedetomidine prevents acute kidney injury after adult cardiac surgery: a meta-analysis of randomized controlled trials. BMC Anesthesiol. 2018;18:7. https://doi.org/10.1186/s12871-018-0472-1
    » https://doi.org/10.1186/s12871-018-0472-1
  • 17
    Sinclair MD. A review of the physiological effects of alpha2-agonists related to the clinical use of medetomidine in small animal practice. Can Vet J. 2003;44(11):885-897.
  • 18
    Villela NR, Nascimento Júnior P, Carvalho LR, Teixeira A. Effects of dexmedetomidine on renal system and on vasopressin plasma levels. Experimental study in dogs. Rev Bras Anestesiol. 2005;55(4):429-440. https://doi.org/10.1590/s0034-70942005000400007
    » https://doi.org/10.1590/s0034-70942005000400007
  • 19
    Geloen A, Chapelier K, Cividjian A, Dantony E, Rabilloud M, May CN, Quintin L. Clonidine and dexmedetomidine increase the pressor response to norepinephrine in experimental sepsis: a pilot study. Crit Care Med. 2013;41(12):e431-8. https://doi.org/10.1097/CCM.0b013e3182986248
    » https://doi.org/10.1097/CCM.0b013e3182986248
  • 20
    Morelli A, Sanfilippo F, Arnemann P, Hessler M, Kampmeier TG, D’Egidio A, Orecchioni A, Santonocito C, Frati G, Greco E, Westphal M, Rehberg SW, Ertmer C. The effect of propofol and dexmedetomidine sedation on norepinephrine requirements in septic shock patients: a crossover trial. Crit Care Med. 2019;47(2):e89-e95. https://doi.org/10.1097/CCM.0000000000003520
    » https://doi.org/10.1097/CCM.0000000000003520
  • 21
    Nelson KM, Patel GP, Hammond DA. Effects from continuous infusions of dexmedetomidine and propofol on hemodynamic stability in critically Ill adult patients with septic shock. J Intensive Care Med. 2020;35(9):875-880. https://doi.org/10.1177/0885066618802269
    » https://doi.org/10.1177/0885066618802269
  • 22
    Miranda ML, Balarini MM, Bouskela E. Dexmedetomidine attenuates the microcirculatory derangements evoked by experimental sepsis. Anesthesiology. 2015;122:619-630. https://doi.org/10.1097/ALN.0000000000000491
    » https://doi.org/10.1097/ALN.0000000000000491
  • 23
    Miyamoto K, Nakashima T, Shima N, Kato S, Ueda K, Kawazoe Y, Ohta Y, Morimoto T, Yamamura H; DESIRE Trial Investigators. Effect of dexmedetomidine on lactate clearance in patients with septic shock: a subanalysis of a multicenter randomized controlled trial. Shock. 2018;50(2):162-166. https://doi.org/10.1097/SHK.0000000000001055
    » https://doi.org/10.1097/SHK.0000000000001055
  • 24
    Gattinoni L, Brazzi L, Pelosi P, Latini R, Tognoni G, Pesenti A, Fumagalli R. A trial of goal-oriented hemodynamic therapy in critically ill patients. SvO2 Collaborative Group. N Engl J Med. 1995;333:1025-1032. https://doi.org/10.1056/NEJM199510193331601
    » https://doi.org/10.1056/NEJM199510193331601
  • 25
    Pascoe PJ. The cardiopulmonary effects of dexmedetomidine infusions in dogs during isoflurane anesthesia. Vet Anaesth Analg. 2015;42(4):360-368. https://doi.org/10.1111/vaa.12220
    » https://doi.org/10.1111/vaa.12220
  • 26
    Memiş D, Hekimoğlu S, Vatan I, Yandim T, Yüksel M, Süt N. Effects of midazolam and dexmedetomidine on inflammatory responses and gastric intramucosal pH to sepsis, in critically ill patients. Br J Anaesth. 2007;98(4):550-552. https://doi.org/10.1093/bja/aem017
    » https://doi.org/10.1093/bja/aem017
  • 27
    Chen H, Sun X, Yang X, Hou Y, Yu X, Wang Y, Wu J, Liu D, Wang H, Yu J, Yi W. Dexmedetomidine reduces ventilator-induced lung injury (VILI) by inhibiting Toll-like receptor 4 (TLR4)/nuclear factor (NF)-κB signaling pathway. Bosn J Basic Med Sci. 2018;18(2):162-169. https://doi.org/10.17305/bjbms.2018.2400
    » https://doi.org/10.17305/bjbms.2018.2400
  • 28
    Yang CL, Chen CH, Tsai PS, Wang TY, Huang CJ. Protective effects of dexmedetomidine-ketamine combination against ventilator-induced lung injury in endotoxemia rats. J Surg Res. 2011;167(2):e273-81. https://doi.org/10.1016/j.jss.2010.02.020
    » https://doi.org/10.1016/j.jss.2010.02.020
  • 29
    Yang CL, Tsai PS, Huang CJ. Effects of dexmedetomidine on regulating pulmonary inflammation in a rat model of ventilator-induced lung injury. Acta Anaesthesiol Taiwan. 2008;46(4):151-159. https://doi.org/10.1016/S1875-4597(09)60002-3
    » https://doi.org/10.1016/S1875-4597(09)60002-3
  • 30
    Calzavacca P, Booth LC, Lankadeva YR, Bailey SR, Burrell LM, Bailey M, Bellomo R, May CN. Effects of clonidine on the cardiovascular, renal, and Inflammatory responses to experimental bacteremia. Shock. 2019;51(3):348-355. https://doi.org/10.1097/SHK.0000000000001134
    » https://doi.org/10.1097/SHK.0000000000001134
  • 31
    Thorgersen EB, Hellerud BC, Nielsen EW, Barratt-Due A, Fure H, Lindstad JK, Pharo A, Fosse E, Tønnessen TI, Johansen HT, Castellheim A, Mollnes TE. CD14 inhibition efficiently attenuates early inflammatory and hemostatic responses in Escherichia coli sepsis in pigs. FASEB J. 2010;24(3):712-722. https://doi.org/10.1096/fj.09-140798
    » https://doi.org/10.1096/fj.09-140798
  • 32
    Taniguchi T, Kurita A, Kobayashi K, Yamamoto K, Inaba H. Dose- and time-related effects of dexmedetomidine on mortality and inflammatory responses to endotoxin-induced shock in rats. J Anesth. 2008;22:221-228. https://doi.org/10.1007/s00540-008-0611-9
    » https://doi.org/10.1007/s00540-008-0611-9
  • 33
    Nishina K, Akamatsu H, Mikawa K, Shiga M, Maekawa N, Obara H, Niwa Y. The effects of clonidine and dexmedetomidine on human neutrophil functions. Anesth Analg. 1999;88(2):452-458. https://doi.org/10.1097/00000539-199902000-00042
    » https://doi.org/10.1097/00000539-199902000-00042
  • 34
    Venn RM, Bryant A, Hall GM, Grounds RM. Effects of dexmedetomidine on adrenocortical function, and the cardiovascular, endocrine and inflammatory responses in post-operative patients needing sedation in the intensive care unit. Br J Anaesth. 2001;86(5):650-656. https://doi.org/10.1093/bja/86.5.650
    » https://doi.org/10.1093/bja/86.5.650

Publication Dates

  • Publication in this collection
    11 Nov 2022
  • Date of issue
    2022

History

  • Received
    12 Mar 2022
  • Reviewed
    09 May 2022
  • Accepted
    13 June 2022
Sociedade Brasileira para o Desenvolvimento da Pesquisa em Cirurgia https://actacirbras.com.br/ - São Paulo - SP - Brazil
E-mail: actacirbras@gmail.com