Skip to main content
Erschienen in: BMC Cardiovascular Disorders 1/2020

Open Access 01.12.2020 | Research article

High plasma levels of pro-inflammatory factors interleukin-17 and interleukin-23 are associated with poor outcome of cardiac-arrest patients: a single center experience

verfasst von: Yu-Gang Zhuang, Yuan-Zhuo Chen, Shu-Qin Zhou, Hu Peng, Yan-Qing Chen, Dong-Jie Li

Erschienen in: BMC Cardiovascular Disorders | Ausgabe 1/2020

Abstract

Background

Systemic inflammation is an important feature of post-cardiac arrest syndrome (PCAS). This study was designed to determine whether the plasma concentrations of some circulating pro-inflammatory cytokines (interleukin-17 [IL-8], IL-22, IL-23 and IL-33) are of value in predicting the outcome of patients after return of spontaneous circulation (ROSC) during the post–cardiac arrest period.

Methods

This was a prospective observational clinical study. In total, 21 patients (survivors, n = 10; non-survivors, n = 11) who experienced cardiac arrest and successful ROSC with expected survival of at least 7 days were consecutively enrolled from January 2016 to December 2017. Of the 21 enrolled patients, ten survived were designated “survivors”. The other eleven patients died between 2 days and 1 months post ROSC. Venous blood was drawn at three time-points: baseline (< 1 h post ROSC), 2 days post ROSC and 7 days post ROSC. Plasma IL-8, IL-22, IL-23 and IL-33 were determined using commercial enzyme-linked immunosorbent assays.

Results

Plasma creatinine levels, but aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels, were elevated in non-survivors compared with survivors. Plasma levels of IL-17, IL-22, IL-23 and IL-33 of the 21 total patients did not change at 2 or 7 days post ROSC compared to baseline. In survivors, the plasma levels of IL-17 and IL-23 at 2 or 7 days post ROSC were lower than baseline. In non-survivors, plasma levels of IL-17 increased compared with baseline. Receiver operating characteristic curve analysis showed that the plasma levels of IL-17 and IL-23 at 2 or 7 days post ROSC were able to predict the mortality of PCAS patients, and positively correlated with Acute Physiology and Chronic Health Evaluation (APACHE)-II score and time to ROSC.

Conclusion

These results provide the first evidence that the elevated plasma IL-17 and IL-23 levels are associated with poor outcome in PCAS patients. The role of IL-17/IL-23 axis post ROSC is worth paying attention to in PCAS patients.

Trial registration

Begleitmaterial
Additional file 1. Plasma levels of IL-23 (Fig. 4D) did to correlate with the time to ROSC. Similar results were oberved in plasma levels of IL-22 and IL-33 (Supplemental Fig. 2).
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12872-020-01451-y.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
APACHE
Acute physiology and chronic health evaluation
AUC
Area under the curve
CPR
Cardiopulmonary resuscitation
IFN
Interferon
IL
Interleukin
PCAS
Post-cardiac arrest syndrome
ROC
Receiver operating characteristic
ROSC
Return of spontaneous circulation
TNF-α
Tumor necrosis factor-α

Background

Despite the increasing understanding on cardiac arrest and development of resuscitation protocols and cardiopulmonary resuscitation (CPR), admission of cardiac arrest patients to the Intensive Care Unit (ICU) is associated with high morbidity and mortality, and consequently great medical resource consumption [1]. Although it has been previously reported that treating the patients following cardiac arrest with induced hypothermia promoted the resuscitation rate and improved neurological outcome [2], this therapy has been challenged by dozens of recent findings [3]. Moreover, even after successful return of spontaneous circulation (ROSC) from cardiac arrest, the survivors would still face significant morbidity and mortality after hospital discharge [4]. This so-called “post-cardiac arrest syndrome” (PCAS) involves four key components: (1) post-cardiac arrest cerebral ischemia injury, (2) post-cardiac arrest myocardial ischemia and malfunction, (3) systemic ischemia/reperfusion injury, and (4) persistent precipitating pathology [4].
Due to the predicament that critical care interventions of PCAS patient is rather expensive and might be ethically problematic if the outcome is poor, predicting the short- and long-term outcomes in PCAS patients becomes a key challenge in the treatment of PCAS patients. To address this problem, discovering novel biomarkers in PACS patients’ blood is valuable in the clinical therapy of PCAS patients [5]. Donnino et al. reported that effective clearance of blood lactate during the early stage was associated with decreased overall mortality in patients with cardiac arrest [6]. In support of this, survivors from cardiac arrest with good neurological outcome had lower lactate levels in the early stage (< 24 h post ROSC) [7], suggesting that early blood lactate level was an independent predictor of survival and neurological outcomes in PCAS patients. Besides, S-100B, an acidic protein with a calcium binding motif produced by astroglial cells in the brain, is a predictive marker for outcome of patients after cardiac arrest [8]. Neuron specific enolase (NSE), another a neural protein primarily produced by neurons and is leaked into the extracellular space after severe injury, is also a useful predictor of outcome in PCAS patients [9]. Lipocalin, might predict the neurological outcomes of PCAS patients, and its predictive value was equivalent to that of NSE [10].
A study conducted by Bro-Jeppesen et al. showed that systemic inflammation triggered by ischemia and multi-organ failure represents a hallmark of the post-cardiac arrest syndrome (PCAS) in patients with out-of-hospital cardiac arrest [11]. In this study, the authors measured level of interleukin-1β (IL-1β), IL-2, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12, IL-13, tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), C-reactive protein, and procalcitonin, and found that levels of IL-6 and IL-10 at baseline were significantly higher in non-survivors compared with survivors [11]. The same group also reported that high IL-6 levels were associated with increased mortality in PCAS patients [12]. Besides, other studies revealed that plasma IL-8 level might be a good marker for predicting the neurological outcome in PCAS patients [8].
In this single-center, case-control study, we measured the plasma levels of IL-17, IL-22, IL-23 and IL-33 in 21 cardiac arrest patients with ROSC at three time-points (baseline [within 1 h post ROSC], 2 days post ROSC and 7 days post ROSC). Although the plasma levels of IL-17, IL-22, IL-23 and IL-33 of the 21 total patients did not change with the course of disease, the plasma levels of IL-17 and IL-23 displayed contrary tendency between the survivors and non-survivors. Moreover, the plasma levels of IL-17 and IL-23 within 48 h post ROSC were potential biomarkers correlating with the neurological injury.

Materials and methods

Patients and ethics approval

A total of 21 adult patients (> 18 years-old) were enrolled to the current prospective observational study. These patients were undergone successful cardiopulmonary resuscitation (CPR) and ROSC, and admitted to our intensive care unit within 1 h after CPR/ROSC at the emergency medical center of Shanghai Tenth People’s Hospital, Shanghai from January 2016 to December 2017. Written informed consent was obtained from every patient. The exclusion criteria include: (1) pregnancy; (2) drowning/hanging; (3) neuroendocrine tumor; (3) traumatic brain injury; (4) sepsis; (6) malignant.
The study was reviewed by the local public school research ethics committee and human-investigation committee for the protection of human subjects in Shanghai Tenth People’s Hospital, Tongji University. Moreover, this investigation was registered in ClinicalTrial.gov (NCT02297776, 2014-9-21, Link: https://​clinicaltrials.​gov/​ct2/​show/​NCT02297776?​term=​02297776&​rank=​1). This study was conducted in compliance with the “Declaration of Helsinki.” Written informed consents were obtained from all participants.

Data collection and blood sampling

All the patients with CA and ROSC were treated with intensive medical care. The Acute Physiology and Chronic Health Evaluation (APACHE)-II score was calculated within 1 h of ICU enrolment. Information about time to ROSC, age, gender, past medical history and the cause of CA were collected. Peripheral venous blood was taken from any easily accessible peripheral vein at three time-points post ROSC: 1 h, 2 days and 7 days. Plasma was obtained [13, 14] and frozen at − 80 °C for further experiment [15].

Kidney and liver function

To monitor the kidney and liver function, the concentrations of creatinine, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in plasma were determined using an Auto Chemistry Analyzer (Hitachi 7180, Tokyo, Japan) as described previously [16, 17].

Enzyme-linked immunosorbent assay (ELISA)

Four kinds of pro-inflammatory factors levels in plasma (IL-17, IL-22, IL-23 and IL-33) using commercial (ELISA) kits as described previously [18, 19]. The ELISA kit for IL-17, IL-22 and IL-23 assays were purchased from R&D Systems (catalogue: HS750, D2000 and D3300 respectively, Minneapolis, MN, USA). The IL-23 assay was purchased from ThermoFisher (catalogue: BMS2023–3, Waltham, MA, USA). The optical density (OD) of each reaction was determined at 450 nM using a TECAN Infinite M200 microplate reader (Tecan, Durham, USA) [2023].

Statistical analysis

All the results were presented as the mean ± SEM. Categorical variables (e.g., gender and comorbidities) are expressed as percentages. ANOVA test or Mann-Whitney U test was used to test differences among groups [24]. Correlation among data was performed by using the Pearson linear test. Receiver-operating characteristic curves were used to evaluate the pro-inflammatory factors as potential markers of CA outcome. Statistical analyses were conducted with GraphPad Prism software suite (version 5.0, La Jolla, CA, USA) [25]. A P-value < 0.05 was considered statistically significant.

Results

Plasma pro-inflammatory factors do not change post ROSC in CA patients

We monitored the plasma levels of pro-inflammatory factors (including IL-17, IL-22, IL-23 and IL-33) with 1 h, two day and seven day post ROSC respectively in the enrolled 21 PCAS patients. However, the four pro-inflammatory factors plasma concentrations did not differ among the three checked time points (Fig. 1A).

Post-ROSC plasma levels of IL-17 and IL-23 have different change patterns between survivors and non-survivors

Then, we grouped the 21 PCAS patients into survivors (n = 10) and non-survivors (n = 11) and analyzed the potential discrepancy between them. The basic features of survivors and non-survivors were illustrated in Table 1. Notably, the plasma IL-17 concentrations in non-survivors at 2 and 7 days post ROSC were significantly higher than those in survivors (Fig. 1B). However, this phenomenon was not observed at 1 h post ROSC (Fig. 1B). Similarly, the plasma IL-23 concentrations in non-survivors were significantly higher than those in survivors at 2 and 7 days post ROSC but not 1 h post ROSC (Fig. 1B). Plasma IL-22 levels post ROSC in both survivors and non-survivors did not change and there was no difference between survivors and non-survivors (Fig. 1B). Also, there was no significant difference in plasma IL-33 levels among 0, 2 and 7 days post ROSC in both survivors and non-survivors and the plasma IL-33 levels did not differ between the survivors and non-survivors (Fig. 1B). There was another interesting phenomenon in plasma IL-17 and IL-23 concentrations that the plasma IL-17 and IL-23 concentrations in survivors declined with time (7 days post ROSC vs 1 h post ROSC, P < 0.05) but not in non-survivors (Fig. 1B).
Table 1
Basic patient characteristics
 
Survivors (n = 10)
Non-survivors (n = 11)
P
Age (years)
74.8
70.7
 
Sex, m/f
6/4
6/4
 
Witnessed by a bystander
9/10
9/11
 
Time to ROSC (min)
6.86
24.5
0.0012
APACHE II score
14.14
29.14
< 0.0001
CPC score
2.28
5
0.0012
Out-of-hospital arrest
2/10
4/11
 
Ventricular defibrillation
2/10
3/11
 
Myocardial infarction
3/10
4/11
 
At presentation in ICU
 Cardiogenic shock
2/10
3/11
 
 Post-resuscitation sepsis
2/10
2/11
 
 Length of ICU stay, days
10.1
16.2
0.0015
 Length of hospital stay, days
19.4
16.3
 
Data are expressed as average or absolute number where appropriate. ROSC, return of spontaneous circulation; ICU, intensive care unit; APACHE II, Acute Physiology and Chronic Health Evaluation II; CPC, cerebral performance category

Renal dysfunction in CA patients post ROSC

We compared the functions of liver (plasma AST and ALT concentrations) and kidney (plasma creatinine concentration) between survivors and non-survivors. As shown in Fig. 2A-B, the plasma AST and ALT levels did not show significant differences between survivors and non-survivors at 0, 2 and 7 days post ROSC. However, the plasma creatinine levels, an index of renal dysfunction, were increased in non-survivors at 2 and 7 days post ROSC (Fig. 2C). There was no association between plasma creatinine level and IL-23/IL-17 levels (data not shown).

Post-ROSC plasma IL-17 and IL-23 levels as possible predictors of survival in PCAS patients

Next, we analyzed whether the plasma levels of IL-17, IL-22, IL-23 and IL-33 might predict mortality. ROC analysis revealed that plasma IL-17 levels at 2 days post ROSC (AUC = 0.8, P = 0.0167) and 7 days post ROSC (AUC = 0.85, P = 0.009) were good predictors of outcome in PCAS patients (Fig. 3A). Plasma IL-23 levels at 2 days (AUC = 0.78, P = 0.0346) or 7 days post ROSC (AUC = 1, P = 0.0002) were also good predictors of outcome in PCAS patients (Fig. 3B). Plasma IL-22 and IL-33 levels were unable to predict the outcome of PCAS patients (Fig. 3C-D).

Post-ROSC plasma IL-17 and IL-23 levels are positively associated with APACHE II score in PCAS patients

To study the relationship between post-ROSC plasma IL-17, IL-22, IL-23 and IL-33 levels and mortality, we performed a linear analysis using APACHE II score, a well-known comprehensive index of severity of neurological deficit [26]. In these PCAS patients, the APACHE II score was positively associated with the plasma IL-17 levels at 2 (r2 = 0.75, P = 0.0001) or 7 days (r2 = 0.53, P = 0.0163) but not 1 h post ROSC (Fig. 4A). A similar association between APACHE II score and plasma IL-23 levels was also observed (Fig. 4B). By contrast, there was no correlation between the plasma levels of IL-22/33 and APACHE II score at the three time-points (Supplemental Fig. 1).

Post-ROSC plasma IL-17 levels are positively associated with time to ROSC in PCAS patients

Time to ROSC is a strong predictor of neurological outcome after ROSC. We found the time to ROSC correlated with the plasma concentrations of IL-17 levels at 2 and 7 days post ROSC (Fig. 4C). Plasma levels of IL-23, IL-22 and IL-33 did to correlate with the time to ROSC (Fig. 4D and Supplemental Fig. 2).

Discussion

As a leading cause of death in developed and many developing countries, cardiac diseases are accounted for around 70% of cases of cardiac arrest; coronary artery diseases are the commonest cause. This is the first study to show the elevated plasma IL-17 and IL-23 levels in PCAS patients and that the plasma concentrations of IL-17 and IL-23 were associated with poor clinical outcome following cardiac arrest. Although the plasma IL-17, IL-22, IL-23 and IL-33 levels did not show any changing trend over time in total PCAS patients, we found significant differences of plasma IL-17 and IL-23 levels between survivors and non-survivors patients at 2 and 7 days post ROSC. By contrary, the plasma IL-17 and IL-23 levels at baseline (< 1 h post ROSC) did not differ between survivors and non-survivors.
It is well-established that the ischemia/reperfusion-induced injury in organs such as heart and brain is closely related to activation of neutrophils/macrophages and extensive releases of pro-inflammatory cytokines and inflammasome [2729]. During this procedure, oxidative stress and endoplasmic reticulum stress also contribute to the cardiac injury [30, 31]. The relationship between inflammation and cardiac arrest has been investigated extensively [32]. Adrie et al. investigated the immune-inflammatory profile in cardiac arrest patients with successfully ROSC and found that high levels of plasma IL-6, IL-8, IL-10, and TNF receptor type II discriminated between survivors and non-survivors [33]. These phenotypes, including the high circulating pro-inflammatory cytokines and the existence of endotoxin in blood, and the absence of ability to product cytokines, mimic the pathophysiology of immunologic and coagulation malfunction observed in sepsis [33]. In agreement with this, TNF-α levels in blood was found to increase during recovery from cardiac arrest in swine model and the increment of TNF-α levels were associated with depression of left ventricle function [34]. Peberdy et al. performed a work to investigate the inflammatory markers including IL-1Ra, IL-6, IL-8, and IL-10 during early stage after ROSC in 102 patients and found baseline IL-6 levels were a good predictor of mortality and strongly associated with in-hospital mortality and poor neurological outcome [35]. Bro-Jeppesen et al. reported baseline plasma IL-6 and IL-10 levels were significantly higher in non-survivors compared with survivors [11, 12]. Melatonin, an anti-oxidant and anti-inflammatory agent, improves neurological outcomes and preserves hippocampal mitochondrial function in cardiac arrest [36], which may be associated with its protection against inflammation [22, 3739], myocardial ischemia-reperfusion injury [4043], ventricular fibrillation [44], cardiac hypertrophy [45], dilated cardiomyopathy [46], and mitochondria-related disorders [47, 48]. As neutralization of IL-17 rescues neuroinflammation [49], melatonin may contribute to the complex role of IL-17 signaling in ROSC [50].
In this study, we selected four pro-inflammatory cytokines (IL-17, IL-22, IL-23 and IL-33) to assess whether they can be used to predict the outcome of PCAS patients. We found plasma IL-17 and IL-23 levels of survivors were significantly lower than those of non-survivors. IL-17 is a major pro-inflammatory cytokine secreted by activated T helper 17 (Th17) and contributes to the pathogenesis of both infectious and inflammatory diseases [51]. Meanwhile, IL-23 acts as a maintainer of Th17 cells and facilitates the expansion of Th17 cells [52]. The key roles of IL-23/IL-17 signaling pathway axis has been well-characterized in arthritis, autoimmune skin diseases, inflammatory bowel disease and neuroinflammation [52]. However, there is no information on IL-23/IL-17 signaling pathway axis in cardiac arrest. In our work, we observed that plasma IL-17 levels decreased over time in survivors but increased in non-survivors during the post-cardiac arrest period. Plasma IL-23 levels also decreased over time in survivors at 2 and 7 days post ROSC although we did observe the increasing of plasma IL-23 level in non-survivors. It should be noted that a large number of previous investigations have documented the critical roles of IL-23/IL-17 signaling in ischemic injury. Li et al. found that innate immune component of kidney ischemic injury requires activation of the IL-23/IL-17 signaling pathways for the neutrophil infiltration [53]. IL-23/IL-17 axis aggravated the left ventricular remodeling after myocardial infarction [54]. In brain ischemia condition, IL-23 seemed to function in the immediate stage of ischemia/reperfusion neural injury, whereas IL-17 played a pivotal role in the delayed phase of ischemia/reperfusion injury [55]. Further, neutralization of the IL-17 axis by an IL-17A–blocking antibody diminished the neutrophil recruitment and pro-inflammatory factors release [56]. These results suggest that IL-17/IL-23 axis may play detrimental effects in ischemic injury. Thus, the decreased plasma IL-17 and IL-23 level in survivors implicate a remarkable reduction of organ injury induced by ischemia, whereas the elevation of IL-17 in non-survivors suggests an aggravation of ischemic injury. It should be noted that several other interleukins such as IL-19 and IL-21 also contribute to myocardial injury during ischemia [5759], so we may need more investigation on them in the future.
Many plasma inflammation markers may predict outcome of patients with cardiac arrest. There were numerous investigations on them. For example, neutrophil lymphocyte ratio, a marker of systemic inflammation, was previously reported to be associated with mortality independently from epinephrine application [60]. Another study showed that neutrophil lymphocyte ratio in survivors was 4.9 (range 0.6–46.5) compared with 8.9 (0.28–96) in non-survivors (P = 0.001) [61]. Blood concentrations of C-reactive protein steadily increased in the several days after ROSC in CA patients and there was a much pronounced higher CRP levels in the patients with system infection [62]. In a logistic regression model, high CRP levels on admission were independently associated with poor neurological outcome at 3 months [62]. However, Oppert et al. found that elevations in procalcitonin but not C-reactive protein are associated with pneumonia after cardiopulmonary resuscitation [63]. Consequently, inflammatory factors are likely to be important mediators and predictors for outcome of cardiac arrest patients. Nevertheless, the concentrations of IL-23 and IL-17, which are crucial to the maintenance of the pro-inflammatory feedback loop by maintaining Th17 cells development and homeostasis, have never been studied in patients with cardiac arrest. Our study is the first to investigate the potential relationship of IL-23/17 axis and cardiac arrest outcome.
Considering that cardiopulmonary resuscitation after cardiac arrest may result in a “sepsis-like” syndrome, the critical involvement of IL-17/IL-23 axis in sepsis may help to understand how IL-17/IL-23 axis modulates the tissue injury in PCAS patients. T cells are primed to promote IL-17 production by increasing STAT3-mediated transcriptional regulation in the lungs of mice with sepsis [64]. Supporting this, blocking IL-17 down-regulated inflammation and conferred protection against sepsis and reduced mortality to both neonatal sepsis and endotoxemia by targeting IL-18 [65]. Additionally, shock mice with Pseudomonas aeruginosa infection showed early and sustained expression of IL-23 in the spleens, and administration of IL-23-neutralizing antibody protected the mice from Gram-negative endotoxic shock [66]. These evidences suggest that IL-17/IL-23 axis substantially contributes to the immunopathology and mortality of sepsis. As a result, IL-17/IL-23 may functions as an inflammation enhancer and the elevation of them in blood indicates a pro-inflammatory status in PCAS patients. Due to the complex role of IL-17/IL-23 in cardiovascular disorders [67], our findings on IL-17/IL-23 axis in PCAS may bring more information about their features in cardiovascular system.
The catecholamine-induced adrenergic receptor stimulation was a common tool to maintain blood pressure in therapy of cardiac arrest, with catecholamine epinephrine being the commonest drug used in ICU [68]. The role of catecholamine and the changed hemodynamics related to the catecholamine levels in cardiac arrest are important characteristics in resuscitation. As early as in 1989, the elevated plasma catecholamines and resuscitation from prolonged cardiac arrest in dogs was firstly reported by Kern et al. [69]. By contrast, Wu et al. showed that the plasma dopamine levels increased, while plasma epinephrine and norepinephrine levels gradually decreased after recurrent ventricular fibrillation in pigs [70]. This suggests that there is still a dispute in plasma catecholamine in cardiac arrest. The relationship between catecholamines and IL-17/23 axis may be an interesting question which may need further investigation in the future.
The influence of cardiac arrest etiology on the differences in IL-17 and IL-23 levels may be another interesting issue. As we have excluded the patients with severe infection, the elevated blood IL-17 and IL-23 were not derived from bacterial infection or LPS stimulation. In fact, almost all the patients included in our study have cardiovascular-metabolic disorders. The cardiovascular diseases, including atherosclerosis, hypertension, myocardial infarction and even atrial fibrillation, seemed to be tightly associated with the activated IL-17/23 axis [52]. Moreover, metabolic disorders, including obesity and diabetes, were also related with IL-17/23 axis [52]. Since these cardiovascular-metabolic dysfunctions were directly associated with IL-17/23 axis, we considered that the etiology of cardiac arrest may have direct relationship with IL-17 and IL-23 levels. Nevertheless, this question may be a complex issue needing in-depth studies which involves more patients.
In summary, we conclude that plasma IL-17/IL-23 axis is a potential predictor of outcome in PCAS patients. Moreover, targeting IL-17/IL-23 axis may serve as a possible therapeutic target to reduce inflammation during post ROSC period in PCAS patients. In order to test these concepts, additional preclinical experiments and clinical trials with larger cohort sizes will be required.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12872-020-01451-y.

Acknowledgements

Not applicable.
The study was approved by the institutional review board of Shanghai Tenth People’s Hospital, Tongji University and registered in ClinicalTrial.​gov (NCT. 02297776). This study was conducted in compliance with the 1964 Helsinki declaration and its later amendments. Written informed consent was obtained from every patient.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis 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.

Publisher’s Note

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

Supplementary information

Additional file 1. Plasma levels of IL-23 (Fig. 4D) did to correlate with the time to ROSC. Similar results were oberved in plasma levels of IL-22 and IL-33 (Supplemental Fig. 2).
Literatur
1.
Zurück zum Zitat Perkins GD, Jacobs IG, Nadkarni VM, Berg RA, Bhanji F, Biarent D, et al. Cardiac arrest and cardiopulmonary resuscitation outcome reports: update of the Utstein Resuscitation Registry Templates for Out-of-Hospital Cardiac Arrest: a statement for healthcare professionals from a task force of the International Liaison Committee on Resuscitation (American Heart Association, European Resuscitation Council, Australian and New Zealand Council on Resuscitation, Heart and Stroke Foundation of Canada, InterAmerican Heart Foundation, Resuscitation Council of Southern Africa, Resuscitation Council of Asia); and the American Heart Association Emergency Cardiovascular Care Committee and the Council on Cardiopulmonary, Critical Care, Perioperative and Resuscitation. Circulation. 2015;132:1286–300.PubMedCrossRef Perkins GD, Jacobs IG, Nadkarni VM, Berg RA, Bhanji F, Biarent D, et al. Cardiac arrest and cardiopulmonary resuscitation outcome reports: update of the Utstein Resuscitation Registry Templates for Out-of-Hospital Cardiac Arrest: a statement for healthcare professionals from a task force of the International Liaison Committee on Resuscitation (American Heart Association, European Resuscitation Council, Australian and New Zealand Council on Resuscitation, Heart and Stroke Foundation of Canada, InterAmerican Heart Foundation, Resuscitation Council of Southern Africa, Resuscitation Council of Asia); and the American Heart Association Emergency Cardiovascular Care Committee and the Council on Cardiopulmonary, Critical Care, Perioperative and Resuscitation. Circulation. 2015;132:1286–300.PubMedCrossRef
2.
Zurück zum Zitat Hypothermia after Cardiac Arrest Study G. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med. 2002;346:549–56.CrossRef Hypothermia after Cardiac Arrest Study G. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med. 2002;346:549–56.CrossRef
3.
Zurück zum Zitat Chan PS, Berg RA. Tang Y, Curtis LH, Spertus JA, American Heart Association's get with the guidelines-resuscitation I. association between therapeutic hypothermia and survival after in-hospital cardiac arrest. JAMA. 2016;316:1375–82.PubMedPubMedCentralCrossRef Chan PS, Berg RA. Tang Y, Curtis LH, Spertus JA, American Heart Association's get with the guidelines-resuscitation I. association between therapeutic hypothermia and survival after in-hospital cardiac arrest. JAMA. 2016;316:1375–82.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Nolan JP, Neumar RW, Adrie C, Aibiki M, Berg RA, Bbttiger BW, et al. Post-cardiac arrest syndrome: epidemiology, pathophysiology, treatment, and prognostication: a scientific statement from the International Liaison Committee on Resuscitation; the American Heart Association Emergency Cardiovascular Care Committee; the Council on Cardiovascular Surgery and Anesthesia; the Council on Cardiopulmonary, Perioperative, and Critical Care; the Council on Clinical Cardiology; the Council on Stroke (Part II). Int Emerg Nurs. 2010;18:8–28.PubMedCrossRef Nolan JP, Neumar RW, Adrie C, Aibiki M, Berg RA, Bbttiger BW, et al. Post-cardiac arrest syndrome: epidemiology, pathophysiology, treatment, and prognostication: a scientific statement from the International Liaison Committee on Resuscitation; the American Heart Association Emergency Cardiovascular Care Committee; the Council on Cardiovascular Surgery and Anesthesia; the Council on Cardiopulmonary, Perioperative, and Critical Care; the Council on Clinical Cardiology; the Council on Stroke (Part II). Int Emerg Nurs. 2010;18:8–28.PubMedCrossRef
5.
Zurück zum Zitat Ujvarosy D, Sebestyen V, Pataki T, Otvos T, Lorincz I, Paragh G, Szabo Z. Cardiovascular risk factors differently affect the survival of patients undergoing manual or mechanical resuscitation. BMC Cardiovasc Disord. 2018;18:227.PubMedPubMedCentralCrossRef Ujvarosy D, Sebestyen V, Pataki T, Otvos T, Lorincz I, Paragh G, Szabo Z. Cardiovascular risk factors differently affect the survival of patients undergoing manual or mechanical resuscitation. BMC Cardiovasc Disord. 2018;18:227.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Donnino MW, Miller J, Goyal N, Loomba M, Sankey SS, Dolcourt B, Sherwin R, Otero R, Wira C. Effective lactate clearance is associated with improved outcome in post-cardiac arrest patients. Resuscitation. 2007;75:229–34.PubMedCrossRef Donnino MW, Miller J, Goyal N, Loomba M, Sankey SS, Dolcourt B, Sherwin R, Otero R, Wira C. Effective lactate clearance is associated with improved outcome in post-cardiac arrest patients. Resuscitation. 2007;75:229–34.PubMedCrossRef
7.
Zurück zum Zitat Donnino MW, Andersen LW, Giberson T, Gaieski DF, Abella BS, Peberdy MA, Rittenberger JC, Callaway CW, Ornato J, Clore J, Grossestreuer A, Salciccioli J, Cocchi MN. National Post-Arrest Research C. initial lactate and lactate change in post-cardiac arrest: a multicenter validation study. Crit Care Med. 2014;42:1804–11.PubMedPubMedCentralCrossRef Donnino MW, Andersen LW, Giberson T, Gaieski DF, Abella BS, Peberdy MA, Rittenberger JC, Callaway CW, Ornato J, Clore J, Grossestreuer A, Salciccioli J, Cocchi MN. National Post-Arrest Research C. initial lactate and lactate change in post-cardiac arrest: a multicenter validation study. Crit Care Med. 2014;42:1804–11.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Mussack T, Biberthaler P, Kanz KG, Wiedemann E, Gippner-Steppert C, Mutschler W, Jochum M. Serum S-100B and interleukin-8 as predictive markers for comparative neurologic outcome analysis of patients after cardiac arrest and severe traumatic brain injury. Crit Care Med. 2002;30:2669–74.PubMedCrossRef Mussack T, Biberthaler P, Kanz KG, Wiedemann E, Gippner-Steppert C, Mutschler W, Jochum M. Serum S-100B and interleukin-8 as predictive markers for comparative neurologic outcome analysis of patients after cardiac arrest and severe traumatic brain injury. Crit Care Med. 2002;30:2669–74.PubMedCrossRef
9.
Zurück zum Zitat Streitberger KJ, Leithner C, Wattenberg M, Tonner PH, Hasslacher J, Joannidis M, Pellis T, Di Luca E, Fodisch M, Krannich A, Ploner CJ, Storm C. Neuron-specific Enolase predicts poor outcome after cardiac arrest and targeted temperature management: a multicenter study on 1,053 patients. Crit Care Med. 2017;45:1145–51.PubMedCrossRef Streitberger KJ, Leithner C, Wattenberg M, Tonner PH, Hasslacher J, Joannidis M, Pellis T, Di Luca E, Fodisch M, Krannich A, Ploner CJ, Storm C. Neuron-specific Enolase predicts poor outcome after cardiac arrest and targeted temperature management: a multicenter study on 1,053 patients. Crit Care Med. 2017;45:1145–51.PubMedCrossRef
10.
Zurück zum Zitat Kaneko T, Fujita M, Ogino Y, Yamamoto T, Tsuruta R, Kasaoka S. Serum neutrophil gelatinase-associated lipocalin levels predict the neurological outcomes of out-of-hospital cardiac arrest victims. BMC Cardiovasc Disord. 2017;17:111.PubMedPubMedCentralCrossRef Kaneko T, Fujita M, Ogino Y, Yamamoto T, Tsuruta R, Kasaoka S. Serum neutrophil gelatinase-associated lipocalin levels predict the neurological outcomes of out-of-hospital cardiac arrest victims. BMC Cardiovasc Disord. 2017;17:111.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Bro-Jeppesen J, Kjaergaard J, Wanscher M, Nielsen N, Friberg H, Bjerre M, Hassager C. Systemic inflammatory response and potential prognostic implications after out-of-hospital cardiac arrest: a substudy of the target temperature management trial. Crit Care Med. 2015;43:1223–32.PubMedCrossRef Bro-Jeppesen J, Kjaergaard J, Wanscher M, Nielsen N, Friberg H, Bjerre M, Hassager C. Systemic inflammatory response and potential prognostic implications after out-of-hospital cardiac arrest: a substudy of the target temperature management trial. Crit Care Med. 2015;43:1223–32.PubMedCrossRef
12.
Zurück zum Zitat Bro-Jeppesen J, Kjaergaard J, Stammet P, Wise MP, Hovdenes J, Aneman A, et al. Predictive value of interleukin-6 in post-cardiac arrest patients treated with targeted temperature management at 33 degrees C or 36 degrees C. Resuscitation. 2016;98:1–8.PubMedCrossRef Bro-Jeppesen J, Kjaergaard J, Stammet P, Wise MP, Hovdenes J, Aneman A, et al. Predictive value of interleukin-6 in post-cardiac arrest patients treated with targeted temperature management at 33 degrees C or 36 degrees C. Resuscitation. 2016;98:1–8.PubMedCrossRef
13.
Zurück zum Zitat Chen S, Wang Z, Xu B, Mi X, Sun W, Quan N, Wang L, Chen X, Liu Q, Zheng Y, Leng J, Li J. The modulation of cardiac contractile function by the pharmacological and toxicological effects of Urocortin2. Toxicol Sci. 2015;148:581–93.PubMedPubMedCentralCrossRef Chen S, Wang Z, Xu B, Mi X, Sun W, Quan N, Wang L, Chen X, Liu Q, Zheng Y, Leng J, Li J. The modulation of cardiac contractile function by the pharmacological and toxicological effects of Urocortin2. Toxicol Sci. 2015;148:581–93.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Li C, Gao Y, Tian J, Xing Y, Zhu H, Shen J. Long-term oral Asperosaponin VI attenuates cardiac dysfunction, myocardial fibrosis in a rat model of chronic myocardial infarction. Food Chem Toxicol. 2012;50:1432–8.PubMedCrossRef Li C, Gao Y, Tian J, Xing Y, Zhu H, Shen J. Long-term oral Asperosaponin VI attenuates cardiac dysfunction, myocardial fibrosis in a rat model of chronic myocardial infarction. Food Chem Toxicol. 2012;50:1432–8.PubMedCrossRef
15.
Zurück zum Zitat Siemian JN, Wang K, Zhang Y, Li JX. Mechanisms of imidazoline I2 receptor agonist-induced antinociception in rats: involvement of monoaminergic neurotransmission. Br J Pharmacol. 2018;175:1519–34.PubMedPubMedCentralCrossRef Siemian JN, Wang K, Zhang Y, Li JX. Mechanisms of imidazoline I2 receptor agonist-induced antinociception in rats: involvement of monoaminergic neurotransmission. Br J Pharmacol. 2018;175:1519–34.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Zhang C, Deng J, Liu D, Tuo X, Xiao L, Lai B, Yao Q, Liu J, Yang H, Wang N. Nuciferine ameliorates hepatic steatosis in high-fat diet/streptozocin-induced diabetic mice through a PPARalpha/PPARgamma coactivator-1alpha pathway. Br J Pharmacol. 2018;175:4218–28.PubMedPubMedCentralCrossRef Zhang C, Deng J, Liu D, Tuo X, Xiao L, Lai B, Yao Q, Liu J, Yang H, Wang N. Nuciferine ameliorates hepatic steatosis in high-fat diet/streptozocin-induced diabetic mice through a PPARalpha/PPARgamma coactivator-1alpha pathway. Br J Pharmacol. 2018;175:4218–28.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Liu J, Jin X, Zhou F, Chen H, Wang W, Liu Y, Wang G, Hao K, Zhang J. Disrupted hepatic pentose phosphate pathway directly participates in and indirectly promotes CYP3A reduction: a new strategy for CYP3A-mediated drug hepatotoxicity. Br J Pharmacol. 2020;177:1538–55.PubMedCrossRef Liu J, Jin X, Zhou F, Chen H, Wang W, Liu Y, Wang G, Hao K, Zhang J. Disrupted hepatic pentose phosphate pathway directly participates in and indirectly promotes CYP3A reduction: a new strategy for CYP3A-mediated drug hepatotoxicity. Br J Pharmacol. 2020;177:1538–55.PubMedCrossRef
18.
Zurück zum Zitat Wei CC, Kong YY, Hua X, Li GQ, Zheng SL, Cheng MH, Wang P, Miao CY. NAD replenishment with nicotinamide mononucleotide protects blood-brain barrier integrity and attenuates delayed tissue plasminogen activator-induced haemorrhagic transformation after cerebral ischaemia. Br J Pharmacol. 2017;174:3823–36.PubMedPubMedCentralCrossRef Wei CC, Kong YY, Hua X, Li GQ, Zheng SL, Cheng MH, Wang P, Miao CY. NAD replenishment with nicotinamide mononucleotide protects blood-brain barrier integrity and attenuates delayed tissue plasminogen activator-induced haemorrhagic transformation after cerebral ischaemia. Br J Pharmacol. 2017;174:3823–36.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Feng L, Sun Y, Song P, Xu L, Wu X, Wu X, Shen Y, Sun Y, Kong L, Wu X, Xu Q. Seselin ameliorates inflammation via targeting Jak2 to suppress the proinflammatory phenotype of macrophages. Br J Pharmacol. 2019;176:317–33.PubMedCrossRef Feng L, Sun Y, Song P, Xu L, Wu X, Wu X, Shen Y, Sun Y, Kong L, Wu X, Xu Q. Seselin ameliorates inflammation via targeting Jak2 to suppress the proinflammatory phenotype of macrophages. Br J Pharmacol. 2019;176:317–33.PubMedCrossRef
20.
Zurück zum Zitat Li DJ, Liu J, Hua X, Fu H, Huang F, Fei YB, Lu WJ, Shen FM, Wang P. Nicotinic acetylcholine receptor alpha7 subunit improves energy homeostasis and inhibits inflammation in nonalcoholic fatty liver disease. Metabolism. 2018;79:52–63.PubMedCrossRef Li DJ, Liu J, Hua X, Fu H, Huang F, Fei YB, Lu WJ, Shen FM, Wang P. Nicotinic acetylcholine receptor alpha7 subunit improves energy homeostasis and inhibits inflammation in nonalcoholic fatty liver disease. Metabolism. 2018;79:52–63.PubMedCrossRef
21.
Zurück zum Zitat Li DJ, Li YH, Yuan HB, Qu LF, Wang P. The novel exercise-induced hormone irisin protects against neuronal injury via activation of the Akt and ERK1/2 signaling pathways and contributes to the neuroprotection of physical exercise in cerebral ischemia. Metabolism. 2017;68:31–42.PubMedCrossRef Li DJ, Li YH, Yuan HB, Qu LF, Wang P. The novel exercise-induced hormone irisin protects against neuronal injury via activation of the Akt and ERK1/2 signaling pathways and contributes to the neuroprotection of physical exercise in cerebral ischemia. Metabolism. 2017;68:31–42.PubMedCrossRef
22.
Zurück zum Zitat Li DJ, Tong J, Li YH, Meng HB, Ji QX, Zhang GY, Zhu JH, Zhang WJ, Zeng FY, Huang G, Hua X, Shen FM, Wang P. Melatonin safeguards against fatty liver by antagonizing TRAFs-mediated ASK1 deubiquitination and stabilization in a beta-arrestin-1 dependent manner. J Pineal Res. 2019;67:e12611.PubMed Li DJ, Tong J, Li YH, Meng HB, Ji QX, Zhang GY, Zhu JH, Zhang WJ, Zeng FY, Huang G, Hua X, Shen FM, Wang P. Melatonin safeguards against fatty liver by antagonizing TRAFs-mediated ASK1 deubiquitination and stabilization in a beta-arrestin-1 dependent manner. J Pineal Res. 2019;67:e12611.PubMed
23.
Zurück zum Zitat Li DJ, Tong J, Zeng FY, Guo M, Li YH, Wang H, Wang P. Nicotinic ACh receptor alpha7 inhibits PDGF-induced migration of vascular smooth muscle cells by activating mitochondrial deacetylase sirtuin 3. Br J Pharmacol. 2019;176:4388–401.PubMedCrossRef Li DJ, Tong J, Zeng FY, Guo M, Li YH, Wang H, Wang P. Nicotinic ACh receptor alpha7 inhibits PDGF-induced migration of vascular smooth muscle cells by activating mitochondrial deacetylase sirtuin 3. Br J Pharmacol. 2019;176:4388–401.PubMedCrossRef
24.
Zurück zum Zitat Lv G, Sun D, Zhang J, Xie X, Wu X, Fang W, Tian J, Yan C, Wang H, Fu F. Lx2-32c, a novel semi-synthetic taxane, exerts antitumor activity against prostate cancer cells in vitro and in vivo. Acta Pharm Sin B. 2017;7:52–8.PubMedCrossRef Lv G, Sun D, Zhang J, Xie X, Wu X, Fang W, Tian J, Yan C, Wang H, Fu F. Lx2-32c, a novel semi-synthetic taxane, exerts antitumor activity against prostate cancer cells in vitro and in vivo. Acta Pharm Sin B. 2017;7:52–8.PubMedCrossRef
25.
Zurück zum Zitat He Y, Zhang W, Guo T, Zhang G, Qin W, Zhang L, Wang C, Zhu W, Yang M, Hu X, Singh V, Wu L, Gref R, Zhang J. Drug nanoclusters formed in confined nano-cages of CD-MOF: dramatic enhancement of solubility and bioavailability of azilsartan. Acta Pharm Sin B. 2019;9:97–106.PubMedCrossRef He Y, Zhang W, Guo T, Zhang G, Qin W, Zhang L, Wang C, Zhu W, Yang M, Hu X, Singh V, Wu L, Gref R, Zhang J. Drug nanoclusters formed in confined nano-cages of CD-MOF: dramatic enhancement of solubility and bioavailability of azilsartan. Acta Pharm Sin B. 2019;9:97–106.PubMedCrossRef
26.
Zurück zum Zitat Donnino MW, Salciccioli JD, Dejam A, Giberson T, Giberson B, Cristia C, Gautam S, Cocchi MN. APACHE II scoring to predict outcome in post-cardiac arrest. Resuscitation. 2013;84:651–6.PubMedCrossRef Donnino MW, Salciccioli JD, Dejam A, Giberson T, Giberson B, Cristia C, Gautam S, Cocchi MN. APACHE II scoring to predict outcome in post-cardiac arrest. Resuscitation. 2013;84:651–6.PubMedCrossRef
27.
28.
Zurück zum Zitat Chen Y, Zhang QS, Shao QH, Wang S, Yuan YH, Chen NH, Wang HB. NLRP3 inflammasome pathway is involved in olfactory bulb pathological alteration induced by MPTP. Acta Pharmacol Sin. 2019;40:991–8.PubMedCrossRef Chen Y, Zhang QS, Shao QH, Wang S, Yuan YH, Chen NH, Wang HB. NLRP3 inflammasome pathway is involved in olfactory bulb pathological alteration induced by MPTP. Acta Pharmacol Sin. 2019;40:991–8.PubMedCrossRef
29.
Zurück zum Zitat Wang S, Yuan YH, Chen NH, Wang HB. The mechanisms of NLRP3 inflammasome/pyroptosis activation and their role in Parkinson's disease. Int Immunopharmacol. 2019;67:458–64.PubMedCrossRef Wang S, Yuan YH, Chen NH, Wang HB. The mechanisms of NLRP3 inflammasome/pyroptosis activation and their role in Parkinson's disease. Int Immunopharmacol. 2019;67:458–64.PubMedCrossRef
30.
Zurück zum Zitat Chen Q, Wang Q, Zhu J, Xiao Q, Zhang L. Reactive oxygen species: key regulators in vascular health and diseases. Br J Pharmacol. 2018;175:1279–92.PubMedCrossRef Chen Q, Wang Q, Zhu J, Xiao Q, Zhang L. Reactive oxygen species: key regulators in vascular health and diseases. Br J Pharmacol. 2018;175:1279–92.PubMedCrossRef
31.
Zurück zum Zitat Wang S, Binder P, Fang Q, Wang Z, Xiao W, Liu W, Wang X. Endoplasmic reticulum stress in the heart: insights into mechanisms and drug targets. Br J Pharmacol. 2018;175:1293–304.PubMedCrossRef Wang S, Binder P, Fang Q, Wang Z, Xiao W, Liu W, Wang X. Endoplasmic reticulum stress in the heart: insights into mechanisms and drug targets. Br J Pharmacol. 2018;175:1293–304.PubMedCrossRef
32.
Zurück zum Zitat Huang S, Frangogiannis NG. Anti-inflammatory therapies in myocardial infarction: failures, hopes and challenges. Br J Pharmacol. 2018;175:1377–400.PubMedPubMedCentralCrossRef Huang S, Frangogiannis NG. Anti-inflammatory therapies in myocardial infarction: failures, hopes and challenges. Br J Pharmacol. 2018;175:1377–400.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Adrie C, Adib-Conquy M, Laurent I, Monchi M, Vinsonneau C, Fitting C, Fraisse F, Dinh-Xuan AT, Carli P, Spaulding C, Dhainaut JF, Cavaillon JM. Successful cardiopulmonary resuscitation after cardiac arrest as a "sepsis-like" syndrome. Circulation. 2002;106:562–8.PubMedCrossRef Adrie C, Adib-Conquy M, Laurent I, Monchi M, Vinsonneau C, Fitting C, Fraisse F, Dinh-Xuan AT, Carli P, Spaulding C, Dhainaut JF, Cavaillon JM. Successful cardiopulmonary resuscitation after cardiac arrest as a "sepsis-like" syndrome. Circulation. 2002;106:562–8.PubMedCrossRef
34.
Zurück zum Zitat Niemann JT, Rosborough JP, Youngquist S, Shah AP, Lewis RJ, Phan QT, Filler SG. Cardiac function and the proinflammatory cytokine response after recovery from cardiac arrest in swine. J Interf Cytokine Res. 2009;29:749–58.CrossRef Niemann JT, Rosborough JP, Youngquist S, Shah AP, Lewis RJ, Phan QT, Filler SG. Cardiac function and the proinflammatory cytokine response after recovery from cardiac arrest in swine. J Interf Cytokine Res. 2009;29:749–58.CrossRef
35.
Zurück zum Zitat Peberdy MA, Andersen LW, Abbate A, Thacker LR, Gaieski D, Abella BS, Grossestreuer AV, Rittenberger JC, Clore J, Ornato J, Cocchi MN, Callaway C, Donnino M. National Post Arrest Research Consortium I. inflammatory markers following resuscitation from out-of-hospital cardiac arrest-a prospective multicenter observational study. Resuscitation. 2016;103:117–24.PubMedCrossRef Peberdy MA, Andersen LW, Abbate A, Thacker LR, Gaieski D, Abella BS, Grossestreuer AV, Rittenberger JC, Clore J, Ornato J, Cocchi MN, Callaway C, Donnino M. National Post Arrest Research Consortium I. inflammatory markers following resuscitation from out-of-hospital cardiac arrest-a prospective multicenter observational study. Resuscitation. 2016;103:117–24.PubMedCrossRef
36.
Zurück zum Zitat Yang L, Wang J, Deng Y, Gong C, Li Q, Chen Q, Li H, Jiang C, Zhou R, Hai K, Wu W, Li T. Melatonin improves neurological outcomes and preserves hippocampal mitochondrial function in a rat model of cardiac arrest. PLoS One. 2018;13:e0207098.PubMedPubMedCentralCrossRef Yang L, Wang J, Deng Y, Gong C, Li Q, Chen Q, Li H, Jiang C, Zhou R, Hai K, Wu W, Li T. Melatonin improves neurological outcomes and preserves hippocampal mitochondrial function in a rat model of cardiac arrest. PLoS One. 2018;13:e0207098.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Schilperoort M, van den Berg R, Bosmans LA, van Os BW, Dolle MET, Smits NAM, et al. Disruption of circadian rhythm by alternating light-dark cycles aggravates atherosclerosis development in APOE*3-Leiden.CETP mice. J Pineal Res. 2020;68:e12614.PubMedCrossRef Schilperoort M, van den Berg R, Bosmans LA, van Os BW, Dolle MET, Smits NAM, et al. Disruption of circadian rhythm by alternating light-dark cycles aggravates atherosclerosis development in APOE*3-Leiden.CETP mice. J Pineal Res. 2020;68:e12614.PubMedCrossRef
38.
Zurück zum Zitat Zhou H, Li D, Zhu P, Ma Q, Toan S, Wang J, Hu S, Chen Y, Zhang Y. Inhibitory effect of melatonin on necroptosis via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates cardiac microvascular ischemia-reperfusion injury. J Pineal Res. 2018;65:e12503.PubMedCrossRef Zhou H, Li D, Zhu P, Ma Q, Toan S, Wang J, Hu S, Chen Y, Zhang Y. Inhibitory effect of melatonin on necroptosis via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates cardiac microvascular ischemia-reperfusion injury. J Pineal Res. 2018;65:e12503.PubMedCrossRef
39.
Zurück zum Zitat Huang CC, Chiou CH, Liu SC, Hu SL, Su CM, Tsai CH, Tang CH. Melatonin attenuates TNF-alpha and IL-1beta expression in synovial fibroblasts and diminishes cartilage degradation: implications for the treatment of rheumatoid arthritis. J Pineal Res. 2019;66:e12560.PubMedCrossRef Huang CC, Chiou CH, Liu SC, Hu SL, Su CM, Tsai CH, Tang CH. Melatonin attenuates TNF-alpha and IL-1beta expression in synovial fibroblasts and diminishes cartilage degradation: implications for the treatment of rheumatoid arthritis. J Pineal Res. 2019;66:e12560.PubMedCrossRef
40.
Zurück zum Zitat Gul-Kahraman K, Yilmaz-Bozoglan M, Sahna E. Physiological and pharmacological effects of melatonin on remote ischemic perconditioning after myocardial ischemia-reperfusion injury in rats: role of Cybb, Fas, NfkappaB, Irisin signaling pathway. J Pineal Res. 2019;67:e12589.PubMedCrossRef Gul-Kahraman K, Yilmaz-Bozoglan M, Sahna E. Physiological and pharmacological effects of melatonin on remote ischemic perconditioning after myocardial ischemia-reperfusion injury in rats: role of Cybb, Fas, NfkappaB, Irisin signaling pathway. J Pineal Res. 2019;67:e12589.PubMedCrossRef
41.
Zurück zum Zitat Han D, Wang Y, Chen J, Zhang J, Yu P, Zhang R, Li S, Tao B, Wang Y, Qiu Y, Xu M, Gao E, Cao F. Activation of melatonin receptor 2 but not melatonin receptor 1 mediates melatonin-conferred cardioprotection against myocardial ischemia/reperfusion injury. J Pineal Res. 2019;67:e12571.PubMedCrossRef Han D, Wang Y, Chen J, Zhang J, Yu P, Zhang R, Li S, Tao B, Wang Y, Qiu Y, Xu M, Gao E, Cao F. Activation of melatonin receptor 2 but not melatonin receptor 1 mediates melatonin-conferred cardioprotection against myocardial ischemia/reperfusion injury. J Pineal Res. 2019;67:e12571.PubMedCrossRef
42.
Zurück zum Zitat Zhang Y, Wang Y, Xu J, Tian F, Hu S, Chen Y, Fu Z. Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK-OPA1 signaling pathways. J Pineal Res. 2019;66:e12542.PubMedCrossRef Zhang Y, Wang Y, Xu J, Tian F, Hu S, Chen Y, Fu Z. Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK-OPA1 signaling pathways. J Pineal Res. 2019;66:e12542.PubMedCrossRef
43.
Zurück zum Zitat Lochner A, Marais E, Huisamen B. Melatonin and cardioprotection against ischaemia/reperfusion injury: What's new? A review. J Pineal Res. 2018;65:e12490.PubMedCrossRef Lochner A, Marais E, Huisamen B. Melatonin and cardioprotection against ischaemia/reperfusion injury: What's new? A review. J Pineal Res. 2018;65:e12490.PubMedCrossRef
44.
Zurück zum Zitat Prado NJ, Egan Benova T, Diez ER, Knezl V, Liptak B, Ponce Zumino AZ, Llamedo-Soria M, Szeiffova Bacova B, Miatello RM, Tribulova N. Melatonin receptor activation protects against low potassium-induced ventricular fibrillation by preserving action potentials and connexin-43 topology in isolated rat hearts. J Pineal Res. 2019;67:e12605.PubMedCrossRef Prado NJ, Egan Benova T, Diez ER, Knezl V, Liptak B, Ponce Zumino AZ, Llamedo-Soria M, Szeiffova Bacova B, Miatello RM, Tribulova N. Melatonin receptor activation protects against low potassium-induced ventricular fibrillation by preserving action potentials and connexin-43 topology in isolated rat hearts. J Pineal Res. 2019;67:e12605.PubMedCrossRef
45.
Zurück zum Zitat Xu L, Su Y, Zhao Y, Sheng X, Tong R, Ying X, Gao L, Ji Q, Gao Y, Yan Y, Yuan A, Wu F, Lan F, Pu J. Melatonin differentially regulates pathological and physiological cardiac hypertrophy: crucial role of circadian nuclear receptor RORalpha signaling. J Pineal Res. 2019;67:e12579.PubMed Xu L, Su Y, Zhao Y, Sheng X, Tong R, Ying X, Gao L, Ji Q, Gao Y, Yan Y, Yuan A, Wu F, Lan F, Pu J. Melatonin differentially regulates pathological and physiological cardiac hypertrophy: crucial role of circadian nuclear receptor RORalpha signaling. J Pineal Res. 2019;67:e12579.PubMed
46.
Zurück zum Zitat Misaka T, Yoshihisa A, Yokokawa T, Sato T, Oikawa M, Kobayashi A, Yamaki T, Sugimoto K, Kunii H, Nakazato K, Takeishi Y. Plasma levels of melatonin in dilated cardiomyopathy. J Pineal Res. 2019;66:e12564.PubMedPubMedCentralCrossRef Misaka T, Yoshihisa A, Yokokawa T, Sato T, Oikawa M, Kobayashi A, Yamaki T, Sugimoto K, Kunii H, Nakazato K, Takeishi Y. Plasma levels of melatonin in dilated cardiomyopathy. J Pineal Res. 2019;66:e12564.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Ding M, Ning J, Feng N, Li Z, Liu Z, Wang Y, Wang Y, Li X, Huo C, Jia X, Xu R, Fu F, Wang X, Pei J. Dynamin-related protein 1-mediated mitochondrial fission contributes to post-traumatic cardiac dysfunction in rats and the protective effect of melatonin. J Pineal Res. 2018;64. Ding M, Ning J, Feng N, Li Z, Liu Z, Wang Y, Wang Y, Li X, Huo C, Jia X, Xu R, Fu F, Wang X, Pei J. Dynamin-related protein 1-mediated mitochondrial fission contributes to post-traumatic cardiac dysfunction in rats and the protective effect of melatonin. J Pineal Res. 2018;64.
48.
Zurück zum Zitat Ding M, Feng N, Tang D, Feng J, Li Z, Jia M, Liu Z, Gu X, Wang Y, Fu F, Pei J. Melatonin prevents Drp1-mediated mitochondrial fission in diabetic hearts through SIRT1-PGC1alpha pathway. J Pineal Res. 2018;65:e12491.PubMedPubMedCentralCrossRef Ding M, Feng N, Tang D, Feng J, Li Z, Jia M, Liu Z, Gu X, Wang Y, Fu F, Pei J. Melatonin prevents Drp1-mediated mitochondrial fission in diabetic hearts through SIRT1-PGC1alpha pathway. J Pineal Res. 2018;65:e12491.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Cristiano C, Volpicelli F, Lippiello P, Buono B, Raucci F, Piccolo M, Iqbal AJ, Irace C, Miniaci MC, Perrone Capano C, Calignano A, Mascolo N, Maione F. Neutralization of IL-17 rescues amyloid-beta-induced neuroinflammation and memory impairment. Br J Pharmacol. 2019;176:3544–57.PubMedCrossRef Cristiano C, Volpicelli F, Lippiello P, Buono B, Raucci F, Piccolo M, Iqbal AJ, Irace C, Miniaci MC, Perrone Capano C, Calignano A, Mascolo N, Maione F. Neutralization of IL-17 rescues amyloid-beta-induced neuroinflammation and memory impairment. Br J Pharmacol. 2019;176:3544–57.PubMedCrossRef
50.
Zurück zum Zitat Alvarez-Sanchez N, Cruz-Chamorro I, Lopez-Gonzalez A, Utrilla JC, Fernandez-Santos JM, Martinez-Lopez A, Lardone PJ, Guerrero JM, Carrillo-Vico A. Melatonin controls experimental autoimmune encephalomyelitis by altering the T effector/regulatory balance. Brain Behav Immun. 2015;50:101–14.PubMedCrossRef Alvarez-Sanchez N, Cruz-Chamorro I, Lopez-Gonzalez A, Utrilla JC, Fernandez-Santos JM, Martinez-Lopez A, Lardone PJ, Guerrero JM, Carrillo-Vico A. Melatonin controls experimental autoimmune encephalomyelitis by altering the T effector/regulatory balance. Brain Behav Immun. 2015;50:101–14.PubMedCrossRef
51.
Zurück zum Zitat Miossec P, Kolls JK. Targeting IL-17 and TH17 cells in chronic inflammation. Nat Rev Drug Discov. 2012;11:763–76.PubMedCrossRef Miossec P, Kolls JK. Targeting IL-17 and TH17 cells in chronic inflammation. Nat Rev Drug Discov. 2012;11:763–76.PubMedCrossRef
52.
53.
Zurück zum Zitat Li L, Huang L, Vergis AL, Ye H, Bajwa A, Narayan V, Strieter RM, Rosin DL, Okusa MD. IL-17 produced by neutrophils regulates IFN-gamma-mediated neutrophil migration in mouse kidney ischemia-reperfusion injury. J Clin Invest. 2010;120:331–42..PubMedCrossRef Li L, Huang L, Vergis AL, Ye H, Bajwa A, Narayan V, Strieter RM, Rosin DL, Okusa MD. IL-17 produced by neutrophils regulates IFN-gamma-mediated neutrophil migration in mouse kidney ischemia-reperfusion injury. J Clin Invest. 2010;120:331–42..PubMedCrossRef
54.
Zurück zum Zitat Yan X, Shichita T, Katsumata Y, Matsuhashi T, Ito H, Ito K, et al. Deleterious effect of the IL-23/IL-17A axis and gammadeltaT cells on left ventricular remodeling after myocardial infarction. J Am Heart Assoc. 2012;1:e004408.PubMedPubMedCentral Yan X, Shichita T, Katsumata Y, Matsuhashi T, Ito H, Ito K, et al. Deleterious effect of the IL-23/IL-17A axis and gammadeltaT cells on left ventricular remodeling after myocardial infarction. J Am Heart Assoc. 2012;1:e004408.PubMedPubMedCentral
55.
Zurück zum Zitat Shichita T, Sugiyama Y, Ooboshi H, Sugimori H, Nakagawa R, Takada I, Iwaki T, Okada Y, Iida M, Cua DJ, Iwakura Y, Yoshimura A. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15:946–50.PubMedCrossRef Shichita T, Sugiyama Y, Ooboshi H, Sugimori H, Nakagawa R, Takada I, Iwaki T, Okada Y, Iida M, Cua DJ, Iwakura Y, Yoshimura A. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15:946–50.PubMedCrossRef
56.
Zurück zum Zitat Gelderblom M, Weymar A, Bernreuther C, Velden J, Arunachalam P, Steinbach K, et al. Neutralization of the IL-17 axis diminishes neutrophil invasion and protects from ischemic stroke. Blood. 2012;120:3793–802.PubMedCrossRef Gelderblom M, Weymar A, Bernreuther C, Velden J, Arunachalam P, Steinbach K, et al. Neutralization of the IL-17 axis diminishes neutrophil invasion and protects from ischemic stroke. Blood. 2012;120:3793–802.PubMedCrossRef
57.
Zurück zum Zitat Wang K, Wen S, Jiao J, Tang T, Zhao X, Zhang M, et al. IL-21 promotes myocardial ischaemia/reperfusion injury through the modulation of neutrophil infiltration. Br J Pharmacol. 2018;175:1329–43.PubMedCrossRef Wang K, Wen S, Jiao J, Tang T, Zhao X, Zhang M, et al. IL-21 promotes myocardial ischaemia/reperfusion injury through the modulation of neutrophil infiltration. Br J Pharmacol. 2018;175:1329–43.PubMedCrossRef
58.
Zurück zum Zitat An W, Yu Y, Zhang Y, Zhang Z, Yu Y, Zhao X. Exogenous IL-19 attenuates acute ischaemic injury and improves survival in male mice with myocardial infarction. Br J Pharmacol. 2019;176:699–710.PubMedPubMedCentralCrossRef An W, Yu Y, Zhang Y, Zhang Z, Yu Y, Zhao X. Exogenous IL-19 attenuates acute ischaemic injury and improves survival in male mice with myocardial infarction. Br J Pharmacol. 2019;176:699–710.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Guo J, Wang H, Li L, Yuan Y, Shi X, Hou S. Treatment with IL-19 improves locomotor functional recovery after contusion trauma to the spinal cord. Br J Pharmacol. 2018;175:2611–21.PubMedPubMedCentralCrossRef Guo J, Wang H, Li L, Yuan Y, Shi X, Hou S. Treatment with IL-19 improves locomotor functional recovery after contusion trauma to the spinal cord. Br J Pharmacol. 2018;175:2611–21.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Weiser C, Schwameis M, Sterz F, Herkner H, Lang IM, Schwarzinger I, Spiel AO. Mortality in patients resuscitated from out-of-hospital cardiac arrest based on automated blood cell count and neutrophil lymphocyte ratio at admission. Resuscitation. 2017;116:49–55.PubMedCrossRef Weiser C, Schwameis M, Sterz F, Herkner H, Lang IM, Schwarzinger I, Spiel AO. Mortality in patients resuscitated from out-of-hospital cardiac arrest based on automated blood cell count and neutrophil lymphocyte ratio at admission. Resuscitation. 2017;116:49–55.PubMedCrossRef
61.
Zurück zum Zitat Patel VH, Vendittelli P, Garg R, Szpunar S, LaLonde T, Lee J, Rosman H, Mehta RH, Othman H. Neutrophil-lymphocyte ratio: a prognostic tool in patients with in-hospital cardiac arrest. World J Crit Care Med. 2019;8:9–17.PubMedPubMedCentralCrossRef Patel VH, Vendittelli P, Garg R, Szpunar S, LaLonde T, Lee J, Rosman H, Mehta RH, Othman H. Neutrophil-lymphocyte ratio: a prognostic tool in patients with in-hospital cardiac arrest. World J Crit Care Med. 2019;8:9–17.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Dell'anna AM, Bini Viotti J, Beumier M, Orbegozo-Cortes D, Donadello K, Scolletta S, Vincent JL, Taccone FS. C-reactive protein levels after cardiac arrest in patients treated with therapeutic hypothermia. Resuscitation. 2014;85:932–8.PubMedCrossRef Dell'anna AM, Bini Viotti J, Beumier M, Orbegozo-Cortes D, Donadello K, Scolletta S, Vincent JL, Taccone FS. C-reactive protein levels after cardiac arrest in patients treated with therapeutic hypothermia. Resuscitation. 2014;85:932–8.PubMedCrossRef
63.
Zurück zum Zitat Oppert M, Reinicke A, Muller C, Barckow D, Frei U, Eckardt KU. Elevations in procalcitonin but not C-reactive protein are associated with pneumonia after cardiopulmonary resuscitation. Resuscitation. 2002;53:167–70.PubMedCrossRef Oppert M, Reinicke A, Muller C, Barckow D, Frei U, Eckardt KU. Elevations in procalcitonin but not C-reactive protein are associated with pneumonia after cardiopulmonary resuscitation. Resuscitation. 2002;53:167–70.PubMedCrossRef
64.
Zurück zum Zitat Mukherjee S, Allen RM, Lukacs NW, Kunkel SL, Carson WF. STAT3-mediated IL-17 production by postseptic T cells exacerbates viral immunopathology of the lung. Shock. 2012;38:515–23.PubMedPubMedCentralCrossRef Mukherjee S, Allen RM, Lukacs NW, Kunkel SL, Carson WF. STAT3-mediated IL-17 production by postseptic T cells exacerbates viral immunopathology of the lung. Shock. 2012;38:515–23.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Wynn JL, Wilson CS, Hawiger J, Scumpia PO, Marshall AF, Liu JH, et al. Targeting IL-17A attenuates neonatal sepsis mortality induced by IL-18. Proc Natl Acad Sci U S A. 2016;113:E2627–35.PubMedPubMedCentralCrossRef Wynn JL, Wilson CS, Hawiger J, Scumpia PO, Marshall AF, Liu JH, et al. Targeting IL-17A attenuates neonatal sepsis mortality induced by IL-18. Proc Natl Acad Sci U S A. 2016;113:E2627–35.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Belladonna ML, Vacca C, Volpi C, Giampietri A, Fioretti MC, Puccetti P, Grohmann U, Campanile F. IL-23 neutralization protects mice from gram-negative endotoxic shock. Cytokine. 2006;34:161–9.PubMedCrossRef Belladonna ML, Vacca C, Volpi C, Giampietri A, Fioretti MC, Puccetti P, Grohmann U, Campanile F. IL-23 neutralization protects mice from gram-negative endotoxic shock. Cytokine. 2006;34:161–9.PubMedCrossRef
67.
Zurück zum Zitat Wenzel UO, Bode M, Kurts C, Ehmke H. Salt, inflammation, IL-17 and hypertension. Br J Pharmacol. 2019;176:1853–63.PubMedCrossRef Wenzel UO, Bode M, Kurts C, Ehmke H. Salt, inflammation, IL-17 and hypertension. Br J Pharmacol. 2019;176:1853–63.PubMedCrossRef
68.
Zurück zum Zitat Paradis NA, Wenzel V, Southall J. Pressor drugs in the treatment of cardiac arrest. Cardiol Clin 2002;20:61–78, viii. Paradis NA, Wenzel V, Southall J. Pressor drugs in the treatment of cardiac arrest. Cardiol Clin 2002;20:61–78, viii.
69.
Zurück zum Zitat Kern KB, Elchisak MA, Sanders AB, Badylak SF, Tacker WA, Ewy GA. Plasma catecholamines and resuscitation from prolonged cardiac arrest. Crit Care Med. 1989;17:786–91.PubMedCrossRef Kern KB, Elchisak MA, Sanders AB, Badylak SF, Tacker WA, Ewy GA. Plasma catecholamines and resuscitation from prolonged cardiac arrest. Crit Care Med. 1989;17:786–91.PubMedCrossRef
70.
Zurück zum Zitat Wu J, Wang S, Li C. Hemodynamic and catecholamine changes after recurrent ventricular fibrillation. J Emerg Med. 2013;44:543–9.PubMedCrossRef Wu J, Wang S, Li C. Hemodynamic and catecholamine changes after recurrent ventricular fibrillation. J Emerg Med. 2013;44:543–9.PubMedCrossRef
Metadaten
Titel
High plasma levels of pro-inflammatory factors interleukin-17 and interleukin-23 are associated with poor outcome of cardiac-arrest patients: a single center experience
verfasst von
Yu-Gang Zhuang
Yuan-Zhuo Chen
Shu-Qin Zhou
Hu Peng
Yan-Qing Chen
Dong-Jie Li
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Cardiovascular Disorders / Ausgabe 1/2020
Elektronische ISSN: 1471-2261
DOI
https://doi.org/10.1186/s12872-020-01451-y

Weitere Artikel der Ausgabe 1/2020

BMC Cardiovascular Disorders 1/2020 Zur Ausgabe

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.