Skip to main content

Systemic interleukin-6 inhibition ameliorates acute neuropsychiatric phenotypes in a murine model of acute lung injury

Abstract

Acute neuropsychiatric impairments occur in over 70% of patients with acute lung injury. Mechanical ventilation is a well-known precipitant of acute lung injury and is strongly associated with the development of acute delirium and anxiety phenotypes. In prior studies, we demonstrated that IL-6 mediates neuropathological changes in the frontal cortex and hippocampus of animals with mechanical ventilation-induced brain injury; however, the effect of systemic IL-6 inhibition on structural and functional acute neuropsychiatric phenotypes is not known. We hypothesized that a murine model of mechanical ventilation-induced acute lung injury (VILI) would induce neural injury to the amygdala and hippocampus, brain regions that are implicated in diverse neuropsychiatric conditions, and corresponding delirium- and anxiety-like functional impairments. Furthermore, we hypothesized that these structural and functional changes would reverse with systemic IL-6 inhibition. VILI was induced using high tidal volume (35 cc/kg) mechanical ventilation. Cleaved caspase-3 (CC3) expression was quantified as a neural injury marker and found to be significantly increased in the VILI group compared to spontaneously breathing or anesthetized and mechanically ventilated mice with 10 cc/kg tidal volume. VILI mice treated with systemic IL-6 inhibition had significantly reduced amygdalar and hippocampal CC3 expression compared to saline-treated animals and demonstrated amelioration in acute neuropsychiatric behaviors in open field, elevated plus maze, and Y-maze tests. Overall, these data provide evidence of a pathogenic role of systemic IL-6 in mediating structural and functional acute neuropsychiatric symptoms in VILI and provide preclinical justification to assess IL-6 inhibition as a potential intervention to ameliorate acute neuropsychiatric phenotypes following VILI.

Introduction

Acute neuropsychiatric impairments, such as delirium and anxiety, occur in over 70% of patients with acute lung injury [1,2,3]. Acute lung injury is a multi-etiological condition that has recently garnered notoriety as a common complication of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), occurring in over one-third of patients who contract the virus [4]. While mechanical ventilation is frequently used to treat patients with acute lung injury, it is also a well-known precipitant of acute lung injury, i.e., ventilator-induced lung injury (VILI), and an established risk factor for acute neuropsychiatric phenotypes [5, 6].

In prior studies, we demonstrated a key pathological role for systemic interleukin-6 (IL-6) in mediating frontal and hippocampal structural neuronal injury in a murine model of VILI [7]; however, what role this cytokine plays in mediating acute neuropsychiatric phenotypes remain unknown. Although various VILI-associated clinical risk factors, such as duration of mechanical ventilation and use of sedatives, have been associated with acute neuropsychiatric symptoms [3, 8], the specific underlying biological mechanisms are not known. It has been hypothesized that, in the general population, immune-mediated injury to the amygdala and hippocampus plays a central role in the pathogenesis of acute neuropsychiatric states, specifically by the cytokines IL-6, IL-1β, and TNF-α [9,10,11,12,13,14,15,16]. Increased IL-6 signaling has been associated with a diversity of neuropsychiatric impairments including delirium [17,18,19] and is frequently elevated in individuals with acute encephalopathy and VILI [20, 21]. We previously demonstrated elevated cortical IL-6 in response to VILI [7]; however, it is not known whether the acute inflammatory response of VILI contributes to acute neuropsychiatric impairments, such as delirium- and anxiety-like behaviors.

In this study, we hypothesized that a murine model of VILI [22] would demonstrate evidence of immune-mediated structural neural injury as well as functional impairments of the amygdala and hippocampus. Furthermore, as systemic IL-6 inhibition has been demonstrated to ameliorate VILI-induced neural injury [23,24,25,26], we hypothesized that systemic IL-6 inhibition would ameliorate both the neural injury and functional impairments of the amygdala and hippocampus, brain regions that are implicated in the pathogenesis of acute neuropsychiatric impairments.

Material and methods

Mice

A total of fifty-four male or female C57BL/6 mice aged 6–7 months old (Jackson Laboratory) were used in this study. Animals were housed in Cedars-Sinai’s AAALAC accredited animal facility under standard conditions (kept in ventilated cages at approximately 21 °C, 40–70% humidity, a 12-h light/dark cycle, with food and water available to the animals ad libitum). All procedures and behavioral testing were approved by Cedars-Sinai’s IACUC (protocol # IACUC007914) prior to any work commencing and entirely new cohorts of animals were used for this study. Based on preliminary immunohistochemical analysis using CC3 (cleaved caspase-3), the greatest group SD and mean difference were 0.12 and 0.3, respectively. Using these values, a power analysis with one-way ANOVA and Tukey’s post hoc test yielded 0.90% power at the 0.05 significance level with n = 5/group. The average attrition rate across experiments was 5.8%. Attrition was exclusively due to associated complications after extubation, and each instance of mortality, the mouse was replaced in a subsequent experiment.

Model establishment/hypothesis generation

We randomized mice to one of the following groups: mice anesthetized and mechanically ventilated with 35 cc/kg tidal volume (VILI) (n = 6), mice anesthetized and mechanically ventilated with 10 cc/kg tidal volume (n = 6), or spontaneously breathing (SB) mice (n = 6).

Hypothesis validation

To examine the effects of IL-6 inhibition on the neuropathology of brain regions implicated in acute neuropsychiatric impairments, mice were randomized to one of the following groups: VILI + Saline (n = 5), VILI + anti (α)-IL-6 antibody (n = 6), VILI + α-IL-6-receptor (n = 6), or SB controls (n = 5).

VILI model

To induce VILI in mice, we utilized a previously validated model of VILI where we subjected fifty-four mice to high tidal volume (35 cc/kg) mechanical ventilation for two hours [22, 27,28,29]. In the mechanically ventilated mice, severe lung injury is induced by mechanically stretching the alveolar walls. The stretching of the alveolar wall causes cell deformation, endothelial and epithelial layer breakdown, interstitial edema, and produces inflammatory infiltrates that can be measured in the bronchoalveolar lavage fluid (BALF) [28]. Histological evidence of acute lung injury using this model has been demonstrated in our prior publication [7].

To rule out possible effects of anesthesia and mechanical ventilation on neural injury and inflammation, we provided the same anesthesia regimen and mechanically ventilated mice with 10 cc/kg [30,31,32]. Mice subjected to VILI or those mechanically ventilated with 10 cc/kg tidal volume were first anesthetized with intraperitoneal injection of a mix of ketamine (Vedco Inc.) and dexmedetomidine (Pfizer) (75 mg/kg and 0.5 mg/kg, respectively), then orotracheally intubated, and lastly mechanically ventilated using an Inspira volume-controlled small animal ventilator (Harvard Apparatus) in ambient, room air. To induce VILI, mice were subjected to the following mechanical ventilation parameters: a tidal volume of 35 cc/kg at a respiratory rate of 70 breaths per minute with zero positive end-expiratory pressure for a duration of 2 h. The mechanical ventilation parameters for the control 10 cc/kg group were as follows: a tidal volume of 10 cc/kg at a respiratory rate of 105 breaths per minute with zero positive end-expiratory pressure for a duration of 2 h. Subcutaneous saline (0.5 mL) was administered to maintain hydration, and the eyes of mice were protected with a thin coat of Paralube (Dechra) immediately before intubation. During mechanical ventilation, the body temperature of mice was maintained using a 38 °C heating pad (Hallowell EMC). Anesthesia was reversed with atipamezole (1 mg/kg in 100 μL of sterile water), and the mice were allowed to recover in their cages on a heating pad for 4 h before euthanasia followed by tissue collection. Control mice were euthanized together with the VILI mice.

IL-6 inhibition

IL-6 signaling was inhibited by systemic administration of two monoclonal antibodies: one which binds IL-6 peptide (α-IL-6) and another that binds the IL-6 receptor (α-IL-6R) (Bio X Cell α-IL-6, clone MP5-20F3; α-IL-6R, clone 15A7). Each antibody-treated mouse received 200 µg of either antibody as a 500µL intraperitoneal injection of a 0.4 ug/µL solution in saline. The dosing ranged from 7.4–9.4 mg/kg, and the average dose was 8.4 mg/kg. Control mice (VILI + Saline) received 500 µL of saline only. After intraperitoneal administration of the IL-6 inhibitors, mice were then placed back into cages for 2 h to allow maximum serum concentrations to be reached [33] after which VILI was induced as outlined in the VILI Model section above. During ventilation, oxygen saturation was recorded using the MouseOx® Plus (v. 1.6; Starr Life Sciences Corp.) system with the thigh sensor.

Brain isolation and treatment

Mice were deeply anesthetized and perfused with room temperature PBS with 0.5 mM ethylenediaminetetraacetic acid (10 mL). Right hemispheres were collected and fixed by submerging in ice-cold PBS buffered 4% paraformaldehyde (Electron Microscopy Sciences) for 30 min, and then cryo-protected in 4% paraformaldehyde + 30% sucrose at 4 °C for 24 h. Free-floating, 30-μm-thick coronal brain cryosections were prepared and stored at 4 °C in PBS + 0.02% sodium azide until staining. Mouse tissue obtained from an acute ischemic stroke mouse model was used as the positive control for hypoxic neural death [34].

Immunohistochemistry (IHC) and microscopy

Sections were affixed to slides by air drying and subjected to heat-induced epitope retrieval for 10 min in antigen-retrieval solution (pH 6.0; Invitrogen) prior to permeabilization/blocking in 5% BSA + 0.25% Triton X-100 in PBS for 1 h at room temperature. Sections were then incubated at 4 °C overnight with primary antibodies diluted in 1% BSA + 0.01% Triton X-100 in PBS (Ab Diluent) (See Additional file 1: Table S1 for antibody information). After washing, sections were incubated with a combination of the appropriate secondary antibody (Alexa Fluor Plus conjugated; Invitrogen) diluted to 4 µg/mL in Ab Diluent for 1 h at room temperature. After washing, sections were incubated in 0.05% Sudan black B in 70% ethanol for 10 min to reduce tissue autofluorescence. Sections were mounted using ProLong Glass with DAPI (Invitrogen). Negative controls were processed using the same protocol with the omission of the primary antibody to assess non-specific labeling. A Carl Zeiss AxioImager Z.2 epi-fluorescence microscope—equipped with standard filter sets/mercury arch lamp, an Apotome 2.0, and an Axiocam HRm camera—controlled by Zen Blue Pro (version 2.3) software was used to acquire and process images. Images of damage marker (e.g., cleaved caspse-3 (CC3)) staining were acquired with a 10 × objective (NA 0.3, Zeiss) as a 3 × 3 tiled image that encompassed the amygdala and a 5 + 5 tile image that encompassed the hippocampus of each section. Images of cytokines (IL-6, IL-1β, and TNF-α) staining were acquired with the Apotome 2.0 and a 20 × objective (NA 0.8, Zeiss) as a single field, 8 µm z-stacks (1 µm interval), and were analyzed and displayed as maximum intensity projections. All acquisition and display settings are the same between groups, and settings were optimized using the VILI group. All images within a figure are set to the same scale.

CC3 was quantified as a potentially reversible neural injury marker that correlates with functional impairments induced by acute systemic disease processes [16, 22].

Image and statistical analysis

Fiji (ImageJ v. 1.53c) software was used for image analysis and quantitation. Prism 9.0.1 (GraphPad) was used for statistical analysis. Three coronal sections (slices) containing the hippocampus and amygdala were analyzed (one ventral, one mid, and one dorsal) for each animal. For damage marker analysis, two different regions of interest (ROIs) were drawn on tiled images of each section: a ROI around the amygdala, or a ROI encompassing the entire hippocampus (both with an average area of 250 µm2). A threshold was set to exclude background pixels per ROI based on the pixel intensity histogram and the number of positive pixels was measured and then expressed as percent area of the ROI. For cytokine analysis, a single-field z-stack projection from the amygdala and hippocampus was analyzed per section. Background pixels were excluded per field based on the pixel intensity histogram and the intensity of the remaining pixels was used to calculate percent area. Values for each protein from the triplicate sections were averaged to yield one number per animal. Analysis was performed by assessors blinded to group allocation. Statistically significant outliers were determined (ROUT method with a Q = 10%) and excluded, and either one-way analysis of variance (ANOVA) with multiple comparisons (Tukey’s) or unpaired Welch’s t test was used to determine statistical significance between groups. Simple linear regression analyses were utilized to examine relationships between variables. For all figures, quantitative data were expressed as mean ± standard deviation (SD) unless otherwise stated. Differences with p ≤ 0.05 were considered significant (* = p ≤ 0.05, ** = p ≤ 0.01, *** = p ≤ 0.001, **** = p ≤ 0.0001).

Behavioral testing

A cohort of fourteen 6–7-month-old mice were used for behavioral testing. VILI was induced in all animals—7 were treated with α-IL-6 antibody and 7 were treated with saline as described above in VILI model and IL-6 inhibition. Behavioral testing was limited to VILI animals to account for differences in locomotor activity induced by acute lung injury and performed 8 h after inducing VILI to allow for sufficient recovery from anesthetic and acute lung injury effects. Further, behavioral testing was limited to female mice only, informed by our experience that male mice with VILI do not participate with the behavioral testing assays.

Open field

Locomotor activity, delirium-, and anxiety-related emotional behaviors were evaluated by placing mice individually into the center of a clear Plexiglas (40 × 40x40 cm) open-field arena and allowing the animals to explore for 45 min [35,36,37]. Activity in the open field was recorded by a computer-operated camera system. Center distance (the distance traveled in the center of the arena) and center time (the time spent in the center of arena) were analyzed by ANY-maze video tracking software version 7.00 (Stoelting Co., Wood Dale, IL, USA). The open field was divided into a 20 × 20 cm central zone and a surrounding border zone during the analysis. Data were collected over the 45-min test session.

Elevated plus maze

Delirium- and anxiety-like behavior was assessed using elevated plus maze [36, 38, 39]. The elevated plus maze crossed arms were approximately 30 cm above the floor during testing, with open and closed arms crossed perpendicularly to each other. A video camera was placed above the apparatus and ANY-maze video tracking software 7.00 (Stoelting Co., Wood Dale, IL, USA) was used to analyze the behavior. At the start of the behavior, the mouse was placed in the center of the crossed arms facing an open arm and allowed to freely explore the entire maze for 5 min. Percent entries in open arms and percent time spent in the open arms were recorded. The percent entries or time spent in open arm was calculated by dividing total entries or time spent in open arm by sum of entries or time spent in open arms and closed arms.

Y-maze

The Y-maze test was used to assess attentional, cognitive, and short-term memory deficits in VILI mice [36, 40]. Each arm of the maze was labeled as either A, B, or C. In each session, the animal is placed in arm A and allowed to explore the three arms for 5 min. Activity in the Y-maze was recorded by a computer-operated video recording system. Number of arm entries are scored from the recorded video file to calculate the percent alternation. The alternation percentage is calculated by dividing the number of alternations by number of possible triads × 100. The elevated plus maze and Y-maze were cleaned with ethanol solution between animals to eliminate odor traces.

Results

The timeline of the experimental design and representative amygdalar and hippocampal brain regions used for analysis are shown in Fig. 1A. There were no significant differences between sex, age, and weight of the mice between the groups.

Fig. 1
figure 1

Cleaved caspase-3 is significantly elevated in the amygdala and hippocampus of VILI mice. A Schematic of experimental design and representative regions of interest (ROIs) of measured brain regions. B Quantification of lung inflammation (%PMN in BALF). C–D Quantification of cleaved caspase-3 (CC3) percent area in the amygdala and the hippocampus where each dot represents one animal. E Representative amygdalar sections stained for CC3 (positive signal displayed in red) overlaid on DAPI nuclear stain (gray). Magnified regions of the indicated area of amygdala from the micrograph directly above. F Representative hippocampal sections stained for CC3 (positive signal displayed in red) overlaid on DAPI nuclear stain (gray). Quantitative data are expressed in mean ± SD. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001

Hypothesis generation

VILI induces neural injury and neuroinflammation

Figure 1B shows an increase in lung inflammation, measured by the percent of polymorphonuclear (PMN) cells in BALF, in the VILI group compared to controls who were either SB or anesthetized and mechanically ventilated with 10 cc/kg (overall ANOVA p ≤ 0.001). Induction of apoptosis triggers multiple pathways that results in the cleavage of caspase-3 to CC3, which further activates downstream apoptotic pathways, ultimately culminating in fragmentation of the genome and cell death [41]. CC3 expression was significantly elevated in the amygdala and hippocampal brain regions of the VILI group compared to SB or 10 cc/kg control groups (overall ANOVA p ≤ 0.0001 and p = 0.0006 respectively; Fig. 1C–D), but there was no evidence of cell death indicated by the absence of terminal deoxynucleotidyl transferase DUTP nick end labeling (TUNEL)-positive staining in the VILI mice (Additional file 1: Fig. S1). Furthermore, to assess any effects of VILI-induced cerebral hypoxic injury, we analyzed the expression of hypoxia-inducible factor-1α (HIF-1α) protein, a highly sensitive transcription factor that is up-regulated in response to hypoxia. There was no evidence of increased expression of HIF-1α in the amygdala of mice with VILI (Additional file 1: Fig. S1). Representative micrographic images of CC3 staining in the amygdala and hippocampus are shown in Fig. 1E–F.

To assess neural activity and stress, we quantified the expression of c-fos and the chaperone protein heat-shock protein-90 (HSP90), respectively. There was an approximately 1.4-fold increase in both c-fos and HSP90 in the amygdala of VILI mice compared to SB (p = 0.0006 and p ≤ 0.0001, respectively; Fig. 2A); however, there were no differences in expression of these proteins within the hippocampus after induction of VILI (Fig. 2B).

Fig. 2
figure 2

VILI increases amygdalar neuronal activity, neuronal stress response, and both amygdalar and hippocampal IL-6 and TNF-α. A–B Quantified levels of cellular stress response (HSP90) and of neuronal activity (c-fos) within the amygdala and hippocampus. C–H IL-6 and TNF-α are significantly increased in VILI brains compared to SB controls in amygdala but not in the hippocampus, while there is no significant change in IL-1β in both areas. I Representative micrographs stained for IL-6 (positive signal displayed in red); cell nuclei are revealed by DAPI staining (gray). Quantitative data are expressed in mean ± SD. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001

To identify the potential roles of cytokine-mediated changes on VILI-induced brain injury, we measured amygdalar and hippocampal expressions of inflammatory cytokines, IL-6, IL-1β, and TNF-α, which are implicated in the pathogenesis of acute neuropsychiatric impairments [16,17,18, 42,43,44,45,46,47,48,49]. Compared to SB, there was a 2.2-fold increase in IL-6 expression in the amygdala and an approximately 3.8-fold increase in amygdalar TNF-α levels (p ≤ 0.01 and p ≤ 0.01, respectively; Fig. 2C–D). There were no significant differences with IL-1β (Fig. 2E). There was an approximately twofold increase in IL-6 expression in the hippocampus (Fig. 2F); however, this observed increase in IL-6 approached but did not reach significance. Additionally, there were no significant changes in TNF-α or IL-1β in the hippocampus (Fig. 2G–H).

PMNs in the BALF were positively and significantly correlated with CC3, IL-6, and TNF-α expression in the amygdala (r2 = 0.4278/p = 0.0274, r2 = 0.5097/p = 0.0091, & r2 = 0.3806/p = 0.0326, respectively; Fig. 3A), while there was no significant relationship with IL-1β (Additional file 1: Fig. S2A). There were positive and significant associations between percent PMNs in the BALF with CC3 (r2 = 0.3950/p = 0.0286; Fig. 3A), but no significant correlations with IL-6 and TNF-α (Fig. 3B, C) or IL-1β (Additional file 1: Fig. S2B) in the hippocampus.

Fig. 3
figure 3

Lung inflammation, cerebral IL-6, and cerebral TNF-α positively and significantly correlate with both amygdalar and hippocampal CC3. A–B Regression analysis between the percent of PMNs in bronchoalveolar lavage fluid (BALF) with amygdalar and hippocampal CC3, IL-6, or TNF-α. SB group is indicated in blue and VILI in red. Significance (p) and fitness (r2) values are indicated. C Regression analysis demonstrating significant positive relationships between amygdalar CC3 with IL-6 and TNF-α. D Micrographs of amygdalar cortical neurons stained for IL-6 (displayed in green) and CC3 (displayed in red). Magnified areas of individual neurons and glial cells are inset. Note that CC3-positive neurons also co-stain for IL-6 in VILI amygdala. E Regression analysis demonstrates a significant positive relationship between hippocampal CC3 with IL-6 but not with TNF-α

Notable associations were found between brain inflammatory cytokines and CC3 in the amygdala: amygdalar IL-6 or TNF-α were significantly and positively correlated with CC3 (r2 = 0.3525/p = 0.0418 & r2 = 0.7685/p = 0.0002, respectively; Fig. 3C); however, there was no significant correlation between IL-1β and amygdalar CC3 (Additional file 1: Fig. S2A). Furthermore, representative micrographs demonstrated an apparent colocalization between IL-6 and CC3-positive neurons in amygdala (Fig. 3D). A significant relationship between hippocampal CC3 expression and IL-6 was identified (r2 = 0.4809/p = 0.0124; Fig. 3E) but not with TNF-α (r2 = 0.0003/p = 0.9534; Fig. 3E). Additionally, a significant association between CC3 and IL-1β (r2 = 0.5267/p = 0.0115; Additional file 1: Fig. S2B) was found in the hippocampus.

Systemic IL-6 inhibition ameliorates amygdalar and hippocampal CC3

To determine possible pathophysiological implications of IL-6 signaling inhibition on structural phenotypes of acute neuropsychiatric impairment, we examined the effects of inhibition of either IL-6 or IL-6-receptor (IL-6R) in mice subjected to VILI [50,51,52]. Percent of PMNs was significantly increased in all three VILI compared to SB; however, there were no significant differences in percent PMNs or oxygen saturation between the VILI groups (Fig. 4A). Amygdalar and hippocampal CC3 expression in α-IL-6- or α-IL-6R-treated VILI groups were significantly reduced compared to saline-treated VILI mice (Fig. 4B–C). There were corresponding significant changes in amygdalar and hippocampal IL-6 and TNF-α, and a nonsignificant trend toward decreased IL-1β expression in the IL-6 inhibited compared to the saline-treated groups in both brain regions (Fig. 4D–I).

Fig. 4
figure 4

IL-6 inhibition significantly reduces amygdalar or hippocampal CC3 expression. A PMNs in BALF after VILI were significantly different from SB regardless of the three intervention groups (ANOVA, F = 9.861, dF = 21, p = 0.0005). There are no significant differences in oxygen saturations between the three VILI intervention groups; VILI + Saline, VILI + α-IL-6, and VILI + α-IL-6-receptor antibody. B–C Amygdalar (ANOVA, F = 8.738, dF = 21, p = 0.0009) and hippocampal (ANOVA, F = 9.806, dF = 21, p = 0.0005) CC3 expressions are significantly increased in the VILI + Saline group compared to SB control, but significantly reduced in both IL-6 inhibited groups. D–E IL-6 (ANOVA, F = 5.160, dF = 21, p = 0.0095) and TNF-α (ANOVA, F = 5.655, dF = 21, p = 0.0066) are significantly increased in the VILI + Saline group compared to SB control, but significantly reduced in both VILI + IL-6 inhibited groups. F There is no significant difference in percent area of amygdalar IL-1β between all groups. G–I Similar to amygdalar cytokines, IL-6 (ANOVA, F = 8.658, dF = 21, p = 0.0009) and TNF-α (ANOVA, F = 4.210, dF = 21, p = 0.0202) are significantly increased in the VILI + Saline group compared to SB control and significantly reduced in both VILI + IL-6 inhibiter groups; however, there is no difference in the percent area of IL-1β between all groups

Positive associations were found between percent PMNs in the BALF and CC3 expression in the brain in both the amygdalar and hippocampal regions (r2 = 0.1941/p = 0.0401 & r2 = 0.2396/p = 0.0208, respectively, Fig. 5A–B), while no significant correlations were found between inflammatory cytokines in the brain and percent PMNs in the BALF (Additional file 1: Fig. S2C). Additionally, there were significant relationships between amygdalar CC3 and IL-6 (r2 = 0.3423/p = 0.0042), amygdalar CC3 and TNF-α (r2 = 0.2163/p = 0.0388), hippocampal CC3 and IL-6 (r2 = 0.4143/p = 0.0012), and hippocampal CC3 and TNF-α (r2 = 0.4798/p = 0.0004; Fig. 5C–D); however, no significant associations were found between CC3 and IL-1β within the same regions (Additional file 1: Fig. S2D). Because IL-6 upregulates a complex signaling pathway, we also examined amygdalar and hippocampal CC3 expression in mice administered α-IL-6 immediately after the completion of ventilation and noted a partial response (Additional file 1: Fig. S3), indicating that early treatment is necessary for the observed effect.

Fig. 5
figure 5

Lung inflammation, cerebral IL-6, and cerebral TNF-α positively and significantly correlate with amygdalar and hippocampal CC3 after IL-6 inhibition. A Regression analysis between the percent of PMNs in bronchoalveolar lavage fluid with amygdalar CC3. SB group is indicated in blue and VILI + Saline in green, VILI + α-IL-6 in red, and VILI + α-IL-6-receptor in brown. Significance (p) and fitness (r2) values are indicated. B Regression analysis between the percent of PMNs in bronchoalveolar lavage fluid with hippocampal CC3. C–D Regression analysis shows a direct and significant relationship between IL-6 and CC3, and TNF-α and CC3 in amygdala and hippocampus

Systemic IL-6 inhibition ameliorates delirium- and anxiety-like behavioral phenotypes

A total of three behavioral tests were used to assess acute neuropsychiatric behaviors: open field, elevated plus maze, and Y-maze [35,36,37,38, 53]. A schematic illustration of the experimental timeline of the behavioral tests is shown in Fig. 6A. The Open-Field test (Fig. 6B) was used as an assessment of anxiety, avoidance behavior, and negative mood alteration. Decreased time spent and distance covered in the center of the open field can be attributed to cognitive deficits, avoidance of trauma related stimuli, and negative alterations in mood, and are markers of PSTD-like behavior. Compared to VILI mice treated with α-IL-6 antibody, the VILI mice treated with saline spent significantly less time in the center area, indicating increased anxiety, avoidance, and mood alterations (p = 0.032, Fig. 6C). There was also significantly less distance covered in the center by the VILI + Saline group, compared to VILI + α-IL-6 group (p = 0.042, Fig. 6D). These changes were not associated with any differences in locomotor activity as measured by total distance traveled or total immobile time (Additional file 1: Fig. S4A–B). Elevated plus maze (Fig. 6E) was used as an additional assay of anxiety-related behavior [39, 40, 54,55,56]. Compared to the VILI + Saline-treated mice, the VILI + α-IL-6 mice entered more and spent more time in open arms suggesting less anxiety and avoidance like behavior[36] (p = 0.044 and p = 0.003 respectively, Fig. 6F–G). Additionally, Y-maze spontaneous alternation (Fig. 6H) was used to assess change in cognition, specifically short-term memory [57, 58]. Compared to VILI + α-IL-6 mice, VILI + Saline mice exhibited significantly less spontaneous alternations in the first 10 alternations that persisted for the entire five-minute duration of the Y-maze test indicating cognitive and short-term memory deficits (p = 0.045, Fig. 6I).

Fig. 6
figure 6

Systemic IL-6 inhibition significantly improves acute delirium- and anxiety-like neuropsychiatric functional impairments following VILI. A Schema of the apparatus and assessment of behavioral function in the center and peripheral zones of the Open Field. B–C In the open-field behavioral test, VILI + Saline mice spent significantly less time and traveled less distance in the center zone compared to the VILI + α-IL-6 group, indicating more anxiety behavior in the VILI + Saline group compared to the α-IL-6 treated group. D Schema of the elevated plus maze for behavioral assessment in open and closed arms. E–F In the elevated plus maze, the VILI + Saline mice entered and spent significantly less time in the open arms as compared to VILI + α-IL-6 mice, indicating increased anxiety and avoidance behaviors in the VILI + Saline group. G Schema of the Y-maze paradigm. H–I VILI + Saline mice had significantly less spontaneous alternations in the Y-maze compared to the α-IL-6 treated group, indicating impaired short-term memory. Quantitative data are expressed in mean ± SD. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001

Discussion

In this study, we demonstrate that systemic IL-6 inhibition reverses VILI-induced neural injury of the amygdala and hippocampus and associated acute delirium- and anxiety-like neuropsychiatric functional impairments. These data provide a pathophysiological basis for the development of acute neuropsychiatric impairments following VILI and serve as preclinical justification to assess the effects of IL-6 inhibition as a potential therapeutic intervention to mitigate these highly morbid psychiatric sequelae of VILI.

Prior imaging studies have reported evidence of amygdalar and hippocampal atrophy in individuals with neuropsychiatric impairments [59,60,61]. Although our results indeed suggest that ongoing cerebral changes during VILI could lead to neural atrophy of the amygdala and hippocampus, the lack of irreversible neural death, as measured by TUNEL staining in this short-term model of VILI, may indicate a window of opportunity to reverse cerebral injury that underlies acute neuropsychiatric impairments after VILI.

While IL-6 is well known to be up-regulated in VILI [17, 18, 32, 34, 46], prior studies have also implicated increased IL-6 signaling in the pathogenesis of diverse neuropsychiatric impairments including, but not limited to, post-traumatic stress disorder, depression, bipolar disorder, and psychotic disorders, each of which may contribute to the risk of acute exacerbation of VILI-induced neuropsychiatric impairments [16, 62,63,64,65]. IL-6 is also commonly elevated in acute encephalopathy, a clinical syndrome that itself can contribute to the development of anxiety-like states via fragmented memories and impaired cognitive processing of distressing experiences [20, 21]. As such, as a marker of neural changes in the amygdala and hippocampus, IL-6 mediated injury may contribute diffuse risk to several neuropsychiatric states. Our interest in the relationship of IL-6 and acute neuropsychiatric impairments is based on the prevalence of these conditions in the VILI population, the upregulation of IL-6 in both VILI and acute neuropsychiatric conditions, as well as the well-established involvement of the amygdala and hippocampus in the etiology of neuropsychiatric impairments. Overall, IL-6 signaling may represent a molecular pathway that is common to the pathogenesis of VILI, delirium-, and anxiety-like behaviors, and explain why these neuropsychiatric impairments occur so commonly as a sequela of VILI.

The results of this study suggest that the IL-6 trans-signaling pathway mediates acute neuropsychiatric phenotypes after VILI. The IL-6 trans-signaling pathway is implicated in the pathogenesis of various neuropsychiatric conditions, including Alzheimer’s disease, Parkinson’s disease, and acute UTI-induced delirium-like states [16, 46, 66]. In IL-6 trans-signaling, IL-6 complexes with the soluble IL-6 receptor in the peripheral circulation and directly induces neuronal injury via the gp130 protein. The results of our study provide additional support for this pathway in inducing acute neuropsychiatric impairments after VILI as systemic inhibition of IL-6 ameliorated these impairments. These data are consistent with the substantial clinical and epidemiological data that associate increased systemic IL-6 levels with acute neuropsychiatric phenotypes after lung injury while further suggesting a direct pathological role of IL-6 in inducing these impairments. However, it is also possible that VILI induces acute neuropsychiatric phenotypes via altered activation of cannabinoid or tryptophan pathways, which are known to modulate peripheral and central inflammation, serotonin generation, and neuropsychiatric function [67, 68]. Future studies are thus needed to determine whether VILI effects on acute neuropsychiatric functions are mediated via alterations in cannabinoid or tryptophan pathways.

In this study, CC3 was used as a neuronal injury marker after VILI. Caspase-3 is a known mediator of neuronal apoptosis, which when cleaved to CC3 is considered among the final steps leading to cell death [69]. However, additional pathological roles for CC3 have been described in acute brain ischemia and diverse neurodegenerative conditions [70, 71]. For instance, caspase-3 has been hypothesized to contribute to cognitive dysfunction by cleaving presenilin and promoting generation of amyloid-β, a key peptide implicated in the pathogenesis of Alzheimer’s disease. As such, while our study has focused on the role of CC3 as an apoptotic mediator, it is possible that non-apoptotic pathways also mediate acute neuropsychiatric impairments—these putative mechanisms should be explored in future studies.

There are notable limitations of this study to consider. Although the 35 cc/kg tidal volume is a previously published model of VILI-induced neural injury [7], the clinical relevance of this supraphysiological tidal volume may be limited. However, the heterogeneity of the many potential etiologies that could lead to mechanical ventilation and VILI likely renders any one model insufficient to reflect the entire phenotypic spectrum of VILI-induced neuropathology—nevertheless, future studies using alternative models of lung injury are indicated. In contrast, an important strength of our model is the lack of any other precipitating cause of lung injury or concurrent systemic illness that may confound the relationship between VILI and acute neuropsychiatric impairments. This feature of our model allows the data to demonstrate that even a short duration of mechanical ventilation resulting in mild isolated VILI followed by a brief recovery period precipitates structural and functional phenotypes of acute neuropsychiatric impairments. A final limitation is that we performed behavioral assessments in female mice only as male mice subjected to VILI do not participate in the behavioral assessments due to reduced levels of activity despite no significant differences in PMNs in the BAL or CC3 expression in the amygdala or hippocampus (Additional file 1: Fig. S5). While beyond the scope of this current study, future research is ongoing and necessary to understand whether male mice have increased susceptibility to more severe hypoactive delirium-like phenotypes, consistent with human studies, and the underlying biological mechanisms [72, 73].

In summary, this study provides first evidence of potentially reversible systemic IL-6-mediated neural injury in the amygdala and hippocampus with corresponding acute delirium- and anxiety-like neuropsychiatric functional impairments in a murine model of VILI. These findings provide a pathophysiological basis for acute neuropsychiatric impairments following VILI and call for future studies to assess the effects of IL-6 inhibition or modulation of IL-6 signaling pathways to ameliorate these highly morbid neuropsychiatric conditions that are known to commonly occur following VILI.

Availability of data and materials

The data that support the findings of this study are available from the corresponding author upon reasonably request.

Abbreviations

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus-2

VILI:

Ventilator-induced lung injury

IL-6:

Interleukin-6

PMN:

Polymorphonuclear

HIF-1α:

Hypoxia-inducible factor-1α

CC3:

Cleaved caspase-3

SB:

Spontaneously breathing

BALF:

Bronchoalveolar lavage fluid

TUNEL:

Terminal deoxynucleotidyl transferase DUTP nick end labeling

HSP90:

Heat-shock protein-90

IL-6R:

IL-6-receptor

References

  1. Sasannejad C, Ely EW, Lahiri S. Long-term cognitive impairment after acute respiratory distress syndrome: a review of clinical impact and pathophysiological mechanisms. Crit Care. 2019;23(1):352.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hager DN, Dinglas VD, Subhas S, Rowden AM, Neufeld KJ, Bienvenu OJ, Touradji P, Colantuoni E, Reddy DR, Brower RG, et al. Reducing deep sedation and delirium in acute lung injury patients: a quality improvement project. Crit Care Med. 2013;41(6):1435–42.

    Article  PubMed  Google Scholar 

  3. Davydow DS, Desai SV, Needham DM, Bienvenu OJ. Psychiatric morbidity in survivors of the acute respiratory distress syndrome: a systematic review. Psychosom Med. 2008;70(4):512–9.

    Article  PubMed  Google Scholar 

  4. Wu C, Chen X, Cai Y. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China (vol 180, pg 934, 2020). Jama Intern Med. 2020;180(7):1031–1031.

    Article  CAS  Google Scholar 

  5. Girard TD, Shintani AK, Jackson JC, Gordon SM, Pun BT, Henderson MS, Dittus RS, Bernard GR, Ely EW. Risk factors for post-traumatic stress disorder symptoms following critical illness requiring mechanical ventilation: a prospective cohort study. Crit Care. 2007;11(1):R28.

    Article  PubMed  PubMed Central  Google Scholar 

  6. International consensus conferences in intensive care medicine. Ventilator-associated Lung Injury in ARDS. This official conference report was cosponsored by the American Thoracic Society, The European Society of Intensive Care Medicine, and The Societe de Reanimation de Langue Francaise, and was approved by the ATS Board of Directors, July 1999. Am J Respir Crit Care Med. 1999;160(6):2118–24.

    Google Scholar 

  7. Sparrow NA, Anwar F, Covarrubias AE, Rajput PS, Rashid MH, Nisson PL, Gezalian MM, Toossi S, Ayodele MO, Karumanchi SA, et al. Interleukin-6 inhibition reduces neuronal injury in a murine model of ventilator-induced lung injury. Am J Respir Cell Mol Biol. 2021;65(4):403–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wade D, Hardy R, Howell D, Mythen M. Identifying clinical and acute psychological risk factors for PTSD after critical care: a systematic review. Minerva Anestesiol. 2013;79(8):944–63.

    CAS  PubMed  Google Scholar 

  9. Shin LM, Kosslyn SM, McNally RJ, Alpert NM, Thompson WL, Rauch SL, Macklin ML, Pitman RK. Visual imagery and perception in posttraumatic stress disorder—a positron emission tomographic investigation. Arch Gen Psychiat. 1997;54(3):233–41.

    Article  CAS  PubMed  Google Scholar 

  10. Rauch SL, van der Kolk BA, Fisler RE, Alpert NM, Orr SP, Savage CR, Fischman AJ, Jenike MA, Pitman RK. A symptom provocation study of posttraumatic stress disorder using positron emission tomography and script-driven imagery. Arch Gen Psychiat. 1996;53(5):380–7.

    Article  CAS  PubMed  Google Scholar 

  11. Liberzon I, Taylor SF, Amdur R, Jung TD, Chamberlain KR, Minoshima S, Koeppe RA, Fig LM. Brain activation in PTSD in response to trauma-related stimuli. Biol Psychiat. 1999;45(7):817–26.

    Article  CAS  PubMed  Google Scholar 

  12. Wang WW, Wang L, Xu H, Cao CQ, Liu P, Luo S, Duan Q, Ellenbroek B, Zhang XY. Characteristics of pro- and anti-inflammatory cytokines alteration in PTSD patients exposed to a deadly earthquake. J Affect Disord. 2019;248:52–8.

    Article  CAS  PubMed  Google Scholar 

  13. Hori H, Kim Y. Inflammation and post-traumatic stress disorder. Psychiatr Clin Neuros. 2019;73(4):143–53.

    Article  Google Scholar 

  14. Gill JM, Saligan L, Woods S, Page G. PTSD is associated with an excess of inflammatory immune activities. Perspect Psychiatr C. 2009;45(4):262–77.

    Article  Google Scholar 

  15. Speer K, Upton D, Semple S, McKune A. Systemic low-grade inflammation in post-traumatic stress disorder: a systematic review. J Inflamm Res. 2018;11:111–21.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Rashid MH, Sparrow NA, Anwar F, Guidry G, Covarrubias AE, Pang H, Bogguri C, Karumanchi SA, Lahiri S. Interleukin-6 mediates delirium-like phenotypes in a murine model of urinary tract infection. J Neuroinflammation. 2021;18(1):247.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Meduri GU, Annane D, Chrousos GP, Marik PE, Sinclair SE. Activation and regulation of systemic inflammation in ARDS: rationale for prolonged glucocorticoid therapy. Chest. 2009;136(6):1631–43.

    Article  PubMed  Google Scholar 

  18. Antunes AA, Sotomaior VS, Sakamoto KS, de Camargo Neto CP, Martins C, Aguiar LR. Interleukin-6 plasmatic levels in patients with head trauma and intracerebral hemorrhage. Asian J Neurosurg. 2010;5(1):68–77.

    PubMed  PubMed Central  Google Scholar 

  19. Smith RJ, Lachner C, Singh VP, Trivedi S, Khatua B, Cartin-Ceba R. Cytokine profiles in intensive care unit delirium. Acute Crit Care. 2022.

  20. Marra A, Pandharipande PP, Patel MB. Intensive care unit delirium and intensive care unit-related posttraumatic stress disorder. Surg Clin North Am. 2017;97(6):1215–35.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Bulic D, Bennett M, Georgousopoulou EN, Shehabi Y, Pham T, Looi JCL, van Haren FMP. Cognitive and psychosocial outcomes of mechanically ventilated intensive care patients with and without delirium. Ann Intensiv Care. 2020;10(1):104.

    Article  Google Scholar 

  22. Sparrow NA, Anwar F, Covarrubias AE, Rajput PS, Rashid MH, Nisson PL, Gezalian MM, Toossi S, Ayodele MO, Karumanchi SA, et al. IL-6 inhibition reduces neuronal injury in a murine model of ventilator-induced lung injury. Am J Respir Cell Mol Biol. 2021;65(4):403–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Le RQ, Li L, Yuan W, Shord SS, Nie L, Habtemariam BA, Przepiorka D, Farrell AT, Pazdur R. FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome. Oncologist. 2018;23(8):943–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gupta S, Wang W, Hayek SS, Chan L, Mathews KS, Melamed ML, Brenner SK, Leonberg-Yoo A, Schenck EJ, Radbel J, et al. Association between early treatment with tocilizumab and mortality among critically Ill patients with COVID-19. Jama Intern Med. 2021;181(1):41–51.

    Article  CAS  PubMed  Google Scholar 

  25. Salama C, Han J, Yau L, Reiss WG, Kramer B, Neidhart JD, Criner GJ, Kaplan-Lewis E, Baden R, Pandit L, et al. Tocilizumab in patients hospitalized with Covid-19 pneumonia. N Engl J Med. 2021;384(1):20–30.

    Article  CAS  PubMed  Google Scholar 

  26. Investigators R-C, Gordon AC, Mouncey PR, Al-Beidh F, Rowan KM, Nichol AD, Arabi YM, Annane D, Beane A, van Bentum-Puijk W, et al. Interleukin-6 receptor antagonists in critically Ill patients with Covid-19. N Engl J Med. 2021;384(16):1491–502.

    Article  Google Scholar 

  27. Wilson MR, Choudhury S, Goddard ME, O’Dea KP, Nicholson AG, Takata M. High tidal volume upregulates intrapulmonary cytokines in an in vivo mouse model of ventilator-induced lung injury. J Appl Physiol (1985). 2003;95(4):1385–93.

    Article  CAS  Google Scholar 

  28. Matute-Bello G, Frevert CW, Martin TR. Animal models of acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2008;295(3):L379-399.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Bastarache JA, Blackwell TS. Development of animal models for the acute respiratory distress syndrome. Dis Model Mech. 2009;2(5–6):218–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Quilez ME, Fuster G, Villar J, Flores C, Marti-Sistac O, Blanch L, Lopez-Aguilar J. Injurious mechanical ventilation affects neuronal activation in ventilated rats. Crit Care. 2011;15(3):R124.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Gonzalez-Lopez A, Lopez-Alonso I, Aguirre A, Amado-Rodriguez L, Batalla-Solis E, Astudillo A, Tomas-Zapico C, Fueyo A, dos Santos CC, Talbot K, et al. Mechanical ventilation triggers hippocampal apoptosis by vagal and dopaminergic pathways. Am J Respir Crit Care Med. 2013;188(6):693–702.

    Article  CAS  PubMed  Google Scholar 

  32. Chen C, Zhang Z, Chen T, Peng M, Xu X, Wang Y. Prolonged mechanical ventilation-induced neuroinflammation affects postoperative memory dysfunction in surgical mice. Crit Care. 2015;19:159.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Prabhakara R, Harro JM, Leid JG, Keegan AD, Prior ML, Shirtliff ME. Suppression of the inflammatory immune response prevents the development of chronic biofilm infection due to methicillin-resistant Staphylococcus aureus. Infect Immun. 2011;79(12):5010–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rajput PS, Lyden PD, Chen B, Lamb JA, Pereira B, Lamb A, Zhao L, Lei IF, Bai J. Protease activated receptor-1 mediates cytotoxicity during ischemia using in vivo and in vitro models. Neuroscience. 2014;281:229–40.

    Article  CAS  PubMed  Google Scholar 

  35. Gould TD, Dao DT, Kovacsics CE. The open field test. In: Gould TD, editor. Mood and anxiety related phenotypes in mice: characterization using behavioral tests. Totowa, NJ: Humana Press; 2009. p. 1–20.

    Chapter  Google Scholar 

  36. Verbitsky A, Dopfel D, Zhang N. Rodent models of post-traumatic stress disorder: behavioral assessment. Transl Psychiatr. 2020;10(1):132.

    Article  Google Scholar 

  37. Seibenhener ML, Wooten MC. Use of the open field maze to measure locomotor and anxiety-like behavior in mice. J Vis Exp. 2015;96:e52434–e52434.

    Google Scholar 

  38. Walf AA, Frye CA. The use of the elevated plus maze as an assay of anxiety-related behavior in rodents. Nat Protoc. 2007;2(2):322–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. File SE. The interplay of learning and anxiety in the elevated plus-maze. Behav Brain Res. 1993;58(1–2):199–202.

    Article  CAS  PubMed  Google Scholar 

  40. Garcia Y, Esquivel N. Comparison of the response of male BALB/c and C57BL/6 mice in behavioral tasks to evaluate cognitive function. Behav Sci (Basel). 2018;8(1):14.

    Article  Google Scholar 

  41. Zille M, Farr TD, Przesdzing I, Müller J, Sommer C, Dirnagl U, Wunder A. Visualizing cell death in experimental focal cerebral ischemia: promises, problems, and perspectives. J Cereb Blood Flow Metab. 2012;32(2):213–31.

    Article  PubMed  Google Scholar 

  42. Meduri GU, Headley S, Kohler G, Stentz F, Tolley E, Umberger R, Leeper K. Persistent elevation of inflammatory cytokines predicts a poor outcome in ARDS. Plasma IL-1 beta and IL-6 levels are consistent and efficient predictors of outcome over time. Chest. 1995;107(4):1062–73.

    Article  CAS  PubMed  Google Scholar 

  43. Goldman JL, Sammani S, Kempf C, Saadat L, Letsiou E, Wang T, Moreno-Vinasco L, Rizzo AN, Fortman JD, Garcia JG. Pleiotropic effects of interleukin-6 in a “two-hit” murine model of acute respiratory distress syndrome. Pulm Circ. 2014;4(2):280–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Cross LJ, Matthay MA. Biomarkers in acute lung injury: insights into the pathogenesis of acute lung injury. Crit Care Clin. 2011;27(2):355–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yang SH, Gangidine M, Pritts TA, Goodman MD, Lentsch AB. Interleukin 6 mediates neuroinflammation and motor coordination deficits after mild traumatic brain injury and brief hypoxia in mice. Shock. 2013;40(6):471–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Erta M, Quintana A, Hidalgo J. Interleukin-6, a major cytokine in the central nervous system. Int J Biol Sci. 2012;8(9):1254–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hewett SJ, Jackman NA, Claycomb RJ. Interleukin-1β in central nervous system injury and repair. Eur J Neurodegener Dis. 2012;1(2):195–211.

    PubMed  PubMed Central  Google Scholar 

  48. Bickenbach J, Biener I, Czaplik M, Nolte K, Dembinski R, Marx G, Rossaint R, Fries M. Neurological outcome after experimental lung injury. Respir Physiol Neurobiol. 2011;179(2–3):174–80.

    Article  PubMed  Google Scholar 

  49. Sinha P, Matthay MA, Calfee CS. Is a “cytokine storm” relevant to COVID-19? JAMA Intern Med. 2020;180(9):1152–4.

    Article  CAS  PubMed  Google Scholar 

  50. Gubernatorova EO, Gorshkova EA, Polinova AI, Drutskaya MS. IL-6: Relevance for immunopathology of SARS-CoV-2. Cytokine Growth Factor Rev. 2020;53:13–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang C, Wu Z, Li JW, Zhao H, Wang GQ. Cytokine release syndrome in severe COVID-19: interleukin-6 receptor antagonist tocilizumab may be the key to reduce mortality. Int J Antimicrob Agents. 2020;55(5): 105954.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chen X, Zhao B, Qu Y, Chen Y, Xiong J, Feng Y, Men D, Huang Q, Liu Y, Yang B, et al. Detectable serum severe acute respiratory syndrome coronavirus 2 viral load (RNAemia) is closely correlated With drastically elevated interleukin 6 level in critically Ill patients with coronavirus disease 2019. Clin Infect Dis. 2020;71(8):1937–42.

    Article  CAS  PubMed  Google Scholar 

  53. Katz RJ, Roth KA, Carroll BJ. Acute and chronic stress effects on open field activity in the rat: implications for a model of depression. Neurosci Biobehav Rev. 1981;5(2):247–51.

    Article  CAS  PubMed  Google Scholar 

  54. Jacobson-Pick S, Audet MC, McQuaid RJ, Kalvapalle R, Anisman H. Social agonistic distress in male and female mice: changes of behavior and brain monoamine functioning in relation to acute and chronic challenges. PLoS ONE. 2013;8(4): e60133.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Richter-Levin G. Acute and long-term behavioral correlates of underwater trauma–potential relevance to stress and post-stress syndromes. Psychiatr Res. 1998;79(1):73–83.

    Article  CAS  Google Scholar 

  56. Cohen H, Zohar J, Matar MA, Zeev K, Loewenthal U, Richter-Levin G. Setting apart the affected: the use of behavioral criteria in animal models of post traumatic stress disorder. Neuropsychopharmacology. 2004;29(11):1962–70.

    Article  PubMed  Google Scholar 

  57. Garcia Y, Esquivel N. Comparison of the response of male BALB/c and C57BL/6 mice in behavioral tasks to evaluate cognitive function. Behav Sci. 2018;8(1):14.

    Article  PubMed Central  Google Scholar 

  58. Muhie S, Gautam A, Chakraborty N, Hoke A, Meyerhoff J, Hammamieh R, Jett M. Molecular indicators of stress-induced neuroinflammation in a mouse model simulating features of post-traumatic stress disorder. Transl Psychiatr. 2017;7(5): e1135.

    Article  CAS  Google Scholar 

  59. Morey RA, Gold AL, LaBar KS, Beall SK, Brown VM, Haswell CC, Nasser JD, Wagner HR, McCarthy G, Mid-Atlantic MW. Amygdala volume changes in posttraumatic stress disorder in a large case-controlled veterans group. Arch Gen Psychiatr. 2012;69(11):1169–78.

    Article  PubMed  Google Scholar 

  60. Cardenas VA, Samuelson K, Lenoci M, Studholme C, Neylan TC, Marmar CR, Schuff N, Weiner MW. Changes in brain anatomy during the course of posttraumatic stress disorder. Psychiat Res-Neuroim. 2011;193(2):93–100.

    Article  Google Scholar 

  61. Henigsberg N, Kalember P, Petrovic ZK, Secic A. Neuroimaging research in posttraumatic stress disorder—Focus on amygdala, hippocampus and prefrontal cortex. Prog Neuro-Psychoph. 2019;90:37–42.

    Article  Google Scholar 

  62. Gola H, Engler H, Sommershof A, Adenauer H, Kolassa S, Schedlowski M, Groettrup M, Elbert T, Kolassa IT. Posttraumatic stress disorder is associated with an enhanced spontaneous production of pro-inflammatory cytokines by peripheral blood mononuclear cells. Bmc Psychiatr. 2013;13:1–8.

    Article  CAS  Google Scholar 

  63. de Oliveira JF, Wiener CD, Jansen K, Portela LV, Lara DR, Souza LDD, da Silva RA, Moreira FP, Oses JP. Serum levels of interleukins IL-6 and IL-10 in individuals with posttraumatic stress disorder in a population-based sample. Psychiat Res. 2018;260:111–5.

    Article  CAS  Google Scholar 

  64. Tortella-Feliu M, Fullana MA, Perez-Vigil A, Torres X, Chamorro J, Littarelli SA, Solanes A, Ramella-Cravaro V, Vilar A, Gonzalez-Parra JA, et al. Risk factors for posttraumatic stress disorder: an umbrella review of systematic reviews and meta-analyses. Neurosci Biobehav Rev. 2019;107:154–65.

    Article  PubMed  Google Scholar 

  65. Passos IC, Vasconcelos-Moreno MP, Costa LG, Kunz M, Brietzke E, Quevedo J, Salum G, Magalhaes PV, Kapczinski F, Kauer-Sant’Anna M. Inflammatory markers in post-traumatic stress disorder: a systematic review, meta-analysis, and meta-regression. Lancet Psychiatr. 2015;2(11):1002–12.

    Article  Google Scholar 

  66. Campbell IL, Erta M, Lim SL, Frausto R, May U, Rose-John S, Scheller J, Hidalgo J. Trans-signaling is a dominant mechanism for the pathogenic actions of interleukin-6 in the brain. J Neurosci. 2014;34(7):2503–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Banner KH, Igney F, Poll C. TRP channels: emerging targets for respiratory disease. Pharmacol Ther. 2011;130(3):371–84.

    Article  CAS  PubMed  Google Scholar 

  68. Izquierdo-García JL, Naz S, Nin N, Rojas Y, Erazo M, Martínez-Caro L, García A, de Paula M, Fernández-Segoviano P, Casals C, et al. A metabolomic approach to the pathogenesis of ventilator-induced lung injury. Anesthesiology. 2014;120(3):694–702.

    Article  PubMed  CAS  Google Scholar 

  69. Pop C, Salvesen GS. Human caspases: activation, specificity, and regulation. J Biol Chem. 2009;284(33):21777–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Fernando P, Brunette S, Megeney LA. Neural stem cell differentiation is dependent upon endogenous caspase 3 activity. Faseb j. 2005;19(12):1671–3.

    Article  CAS  PubMed  Google Scholar 

  71. Li Z, Sheng M. Caspases in synaptic plasticity. Mol Brain. 2012;5:15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Bowman EML, Cunningham EL, Page VJ, McAuley DF. Phenotypes and subphenotypes of delirium: a review of current categorisations and suggestions for progression. Crit Care. 2021;25(1):334.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Kolanowski AM, Hill NL, Kurum E, Fick DM, Yevchak AM, Mulhall P, Clare L, Valenzuela M. Gender differences in factors associated with delirium severity in older adults with dementia. Arch Psychiatr Nurs. 2014;28(3):187–92.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Funding provided by NIH/NIA R03AG064106 (SL), American Academy of Neurology Institute (SL), F. Widjaja Foundation (SL).

Author information

Authors and Affiliations

Authors

Contributions

SL contributed to the study concept and design. FA, NAS, MHR, GG, and SL contributed to the acquisition of data. NAS, FA, MHR, MK, ID, EWE, SAK, and SL did the analysis and interpretation of data. NAS, FA, MMG, MK, ID, EWE, and SL drafted the manuscript. NAS, FA, and MHR did the statistical analysis. SL did the study supervision. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Shouri Lahiri.

Ethics declarations

Ethical approval and consent to participate

All experiments were conducted in accordance with Cedars-Sinai Medical Center Institutional Animal Care and Use Committee (IACUC) guidelines under an approved protocol and complied with current US law.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1.

Antibodies used for immunohistochemistry. Fig. S1. No evidence of hypoxic ischemic injury in our VILI model demonstrated by the lack of hypoxic ischemic factor-1α (HIF-1 α) staining in the amygdala compared to an acute ischemic stroke positive control model. Fig. S2. Correlations between lung inflammation, amygdalar cytokine concentrations, and CC3. AB include SB and VILI animals only while C–D include animals from SB, VILI+Saline, VILI+α-IL-6, and VILI+α-IL-6R. A No significant correlations between %PMNs in BALF and amygdalar IL-1β or between amygdalar IL-1β and amygdalar CC3. B No significant correlation between %PMNs in BALF and hippocampal IL-1β. Direct and significant correlation between hippocampal IL-1β and hippocampal CC3. C No significant correlations between %PMNs in BALF and amygdalar IL-1β or between amygdalar IL-1β and amygdalar CC3. D No significant correlations between %PMNs in BALF and hippocampal IL-1β or between hippocampal IL-1β and hippocampal CC3. Fig. S3. A partial response to the administration α-IL-6 immediately after the completion of ventilation. Independent sample t tests revealed A no significant difference in amygdalar cleaved caspase-3 (CC3) between saline and α-IL-6 treated males. B However, in the hippocampus there was a trend toward significance, with α-IL-6 treated animals have lower CC3. These results indicate early treatment is necessary for the observed effect in both brain regions. N = 5–6 per group and data are expressed in mean ± SD. *p<0.05. Fig. S4. These data show no significant differences in A total distance traveled or B total time immobile between the VILI+Saline and VILI+α-IL-6 groups, indicating that overall level of activity did not explain the differences in behavioral function between the two groups. Fig. S5. Independent sample t tests indicated no significant differences between male (n = 3) and female (n = 3) mice after ventilation-induced acute lung injury (VILI) for A %PMNs in BALF, B amygdalar cleaved caspase-3 (CC3), and C hippocampal CC3. These results were observed despite male mice not participating in the behavioral assessments due to reduced levels of activity following VILI (results not shown). Data are expressed in mean ± SD.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Anwar, F., Sparrow, N.A., Rashid, M.H. et al. Systemic interleukin-6 inhibition ameliorates acute neuropsychiatric phenotypes in a murine model of acute lung injury. Crit Care 26, 274 (2022). https://doi.org/10.1186/s13054-022-04159-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13054-022-04159-x

Keywords