Skip to main content
Erschienen in: Inflammation 1/2024

Open Access 29.10.2023 | RESEARCH

Interleukin-4 Modulates Neuroinflammation by Inducing Phenotypic Transformation of Microglia Following Subarachnoid Hemorrhage

verfasst von: Jing Wang, Lili Wang, Qingjian Wu, Yichen Cai, Chengfu Cui, Ming Yang, Baoliang Sun, Leilei Mao, Yuan Wang

Erschienen in: Inflammation | Ausgabe 1/2024

Abstract

Neuroinflammation, a key pathological feature following subarachnoid hemorrhage (SAH), can be therapeutically targeted by inhibiting microglia M1 polarization and promoting phenotypic transformation to M2 microglia. Interleukin-4 (IL-4) is a pleiotropic cytokine known to its regulation of physiological functions of the central nervous system (CNS) and mediate neuroinflammatory processes. However, its specific role in neuroinflammation and microglia responses following SAH remains unexplored. In this investigation, we established both in vivo and in vitro SAH models and employed a comprehensive array of assessments, including ELISA, neurofunctional profiling, immunofluorescence staining, qRT-PCR, determination of phagocytic capacity, and RNA-Seq analyses. The findings demonstrate an elevated expression of IL-4 within cerebrospinal fluid (CSF) subsequent to SAH. Furthermore, exogenous administration of IL-4 ameliorates post-SAH neurofunctional deficits, attenuates cellular apoptosis, fosters M2 microglia phenotype conversion, and mitigates neuroinflammatory responses. The RNA-Seq analysis signifies that IL-4 governs the modulation of neuroinflammation in microglia within an in vitro SAH model through intricate cascades of signaling pathways, encompassing interactions between cytokines and cytokine receptors. These discoveries not only augment comprehension of the neuropathogenesis associated with post-SAH neuroinflammation but also present novel therapeutic targets for the management thereof.

Graphical Abstract

Hinweise

Publisher's Note

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

INTRODUCTION

Subarachnoid hemorrhage (SAH), primarily arising from intracranial arterial rupture, is a catastrophic ailment with significant mortality rates, constituting 5% of all stroke cases [1]. Notwithstanding ongoing enhancements in aneurysm management and neuroimaging methodologies, there has been limited alteration in both mortality and morbidity rates, with a substantial 50% of survivors enduring irreversible impairments [24]. The management of SAH and the attainment of favorable functional outcomes persist as formidable challenges.
Neuroinflammation stands as a primary pathological hallmark subsequent to SAH. Following SAH, peripheral immune cells are recruited and activated within damaged tissues, while resident microglia/macrophages undergo activation to initiate the inflammatory cascade [5, 6]. The phenotypes of microglia (M1 and M2 phenotypes) undergo alterations in response to changes in microenvironmental signals, thereby assuming distinct roles [7]. The activated M1 phenotype stimulates elevated levels of pro-inflammatory factors and augments the release of cytotoxic reactive oxygen species (ROS), leading to tissue inflammation and cerebral damage [8]. Conversely, the alternative activated M2 phenotypes release anti-inflammatory cytokines and neurotrophic factors that exert anti-inflammatory effects [9]. Inhibiting microglia M1 polarization while promoting the phenotypic transformation of M2 microglia has proven effective in mitigating acute brain injury, enhancing clinical outcomes [8], and represents a potential therapeutic avenue following SAH.
Interleukin-4 (IL-4), a pleiotropic cytokine, assumes a vital role in modulating the physiological functions of the central nervous system (CNS) and mediating neuroinflammatory processes [10, 11]. The conventional viewpoint suggests that IL-4 is primarily secreted by T helper-like 2 cells. However, emerging research has revealed additional sources, including neurons within ischemic brain tissue and microglial within the CNS themselves, which also contribute to the endogenous defense mechanisms [12, 13]. IL-4 exhibits the ability to modulate immune responses and neuroinflammation, thereby exerting neuroprotective and neurorepair effects in various experimental models of CNS diseases, such as traumatic brain injury, cerebrovascular accident, spinal cord injury, and autoimmune encephalomyelitis [14]. Notably, IL-4 induces a polarized microglia/macrophage phenotype that enhances the clearance of tissues through phagocytosis. Studies have demonstrated that the presence of IL-4, including its exogenous administration, induces the expression of genes characteristic of the M2 microglia phenotype in a focal experimental cerebral ischemia model, subsequently promoting the improvement of neurological function [12]. Early elevation of IL-4 levels in the cerebrospinal fluid of patients with SAH has been found in clinical studies, and IL-4 has been considered to be a protective factor associated with survival [1517]. However, despite its prominent role in regulating inflammatory responses in other CNS disorders, the effects of IL-4 on microglia following SAH have not been investigated.
The present study aimed to examine the role of IL-4 in regulating neuroinflammation and improving neurological outcomes in both in vitro and in vivo models of SAH. Through modulation of microglial cell polarization, IL-4 demonstrated its potential in regulating inflammatory responses. We further validated the potential mechanism of IL-4 to modulate inflammatory responses in microglia in vitro by RNA-Seq analysis, providing novel therapeutic targets for the treatment of neuroinflammation after SAH.

MATERIALS AND METHODS

Animals and SAH Model

A total of 132 adult Sprague–Dawley (SD) rats (male, 260–300 g, 7–8 weeks old) were used in this study. All experimental procedures were reviewed and approved by the Institutional Animal Care and Use Committee of Shandong First Medical University and conformed to the protocols set forth by the National Institutes of Health in the USA (approval No. 2019011).
The SAH model was established using the intravascular puncture technique, as previously described [18, 19]. To summarize, rats were anesthetized with isoflurane (5% for induction and 2.5% for maintenance). The right common carotid artery (CCA) was surgically exposed and the internal carotid artery (ICA) and external carotid artery (ECA) were then isolated from their bifurcations. A nylon wire was inserted into the ICA via the ECA and advanced approximately 21–22 mm from the arterial bifurcation until a distinct sensation of penetration persisted for 15 s. In the sham-operated group, the same procedure was followed, excluding the puncturing of the vessel.

Neurological Function Assessment

Neurologic function was evaluated blindly at 1, 3, and 5 days post-SAH employing a modified Garcia score encompassing six dimensions including spontaneous activity, spontaneous movement of all limbs, forelimb movement, climbing wall of wire cage, reaction to touch on both side of trunk, and response to vibrissae touch. Each dimension was scored on a scale of 0–3 or 1–3, with a minimum total score of 3 and a maximum of 18 [18].
At 5 days after the procedure, the severity of SAH bleeding was evaluated [18]. Briefly, the basal cistern of the rat cranial base was divided into six segments, and each segment was rated on a grade of 0–3 based on the amount of bleeding. The sum of the six regions was the SAH score (0–18), and a score of 0–7 was excluded from the study.
Additionally, the hang wire test was employed to evaluate somatosensory motor function [20]. Briefly, rats were placed on a strip of wire with supports at both ends, and their behavior on the wire within 30 s was recorded for scoring: 0, falling; 1, two front paws hanging from the wire strip; 2, front paws hanging from the wire and attempting to climb up; 3, two front paws and one or two hind paws hanging from the wire strip; 4, four paws hanging from the wire strip with the tail wrapped around the wire strip; and 5, escaping to the supports. The experiment was repeated three times and the average score was taken for calculation.

Experimental Design In Vivo

The experimental design employed is depicted in Fig. 1a. In the in vivo experiments, aimed at investigating the alterations in IL-4 expression levels subsequent to SAH, rats were randomly assigned to two groups: the sham group (n = 6) and the SAH group (n = 36). Peripheral blood and CSF samples were taken for enzyme-linked immunosorbent assay (ELISA) at 12 h, 24 h, 3 days, and 5 days post-SAH.
To examine the impact of IL-4 on neuroinflammation after SAH, rats were randomly divided into four groups: the sham group (administered with phosphate-buffered saline (PBS), n = 19), the sham + IL-4 group (administered with IL-4. n = 14), the SAH group (subjected to SAH and administered with PBS, n = 29), and the SAH + IL-4 group (subjected to SAH and administered with IL-4, n = 28). All rats were euthanized 5 days post-SAH, and subsequent analyses included immunofluorescence staining, ELISA, and quantitative real-time polymerase chain reaction (qRT-PCR).

Enzyme-Linked Immunosorbent Assay

To investigate the alterations in endogenous IL-4 levels following SAH, sham and SAH rats were anesthetized with isoflurane and placed on a stereotaxic apparatus, and CSF samples were collected at the site of the occipital pool [21]. Blood samples were taken by cardiac puncture and then centrifuged for serum (3000 × g for 10 min at 4 °C). IL-4 levels in the CSF and serum samples were assessed using a rat IL-4 ELISA kit, following the manufacturer’s instructions (Mlbio, China).
To assess the impact of IL-4 treatment on the expression levels of inflammatory factors TNF-α, iNOS, and IL-10, brain tissues were obtained from the ipsilateral basal cortex 5 days after SAH and assayed using rat ELISA kits (Mlbio, China). BV2 cell culture supernatants in each group were collected to measure the expression levels of TNF-α, iNOS, and IL-10 through mouse ELISA kits (Mlbio, China). The absorbance density (OD) values were measured at a specific wavelength of 450 nm using a spectrophotometer, and the concentrations of each sample were calculated accordingly.

Administration of IL-4

IL-4 administration was performed as shown previously [22]. After isoflurane anesthesia, animals were placed on a brain stereotaxic instrument and immobilized. According to the in vivo experimental design, an implantable slow-release pump (RWD Life Science, China) containing recombinant rat IL-4 (Peprotech, 60 ng/day) or PBS was positioned into the lateral ventricle of the lesion (coordinates: AP 1.0 mm, ML 1.5 mm, DV 4.5 mm) immediately after the onset of SAH. The pump was then started and the infusion was continued at a constant rate of 0.5 μl/h for 5 days until the rats were euthanized [22].

Immunofluorescence

Immunostaining was conducted following established protocols [23]. Briefly, brain tissues were collected after cardiac perfusion with cold 4% paraformaldehyde (PFA), placed in 4% PFA overnight, and then dehydrated using 30% sucrose/PBS for 4 days. Tissues at distances of − 2.5 to − 5 mm from bregma were selected for serial sections of 25-μm thickness. Sections were incubated overnight with primary antibody, including anti-NeuN (1:200, MAB377, Abcam), anti-cleaved caspase-3 (1:400, #9661, CST), anti-Iba-1 (1:1000, 019–19741, Wako, Japan), anti-Iba-1 (1:500, ab5076, Abcam), anti-CD68 (1:1000, ab125212, Abcam), and anti-CD206 (1:500, AF2535, R&D, USA), and then incubated for 2 h at room temperature with the appropriate fluorescent secondary antibody (Jackson ImmunoResearch Laboratories). DAPI (Southern Biotech) was utilized for nuclear staining and mounting. Micrographs were taken using a confocal microscope (Olympus, Japan). Three sections per rat were used, and three random fields in the ipsilateral basal cortex were acquired in each section, and the immunopositive cells in the basal cortex were quantified using Image J software [24].

Cell Culture and Treatment

In the in vitro experiments, BV2 cells were divided into three groups for cytophagocytosis assay, qRT-PCR, and RNA-Seq analysis: (1) control group, (2) control +IL-4 group, (3) oxyhemoglobin group, and (4) oxyhemoglobin +IL-4 group. The microglial BV2 cells (purchased from BULEFBIO, China) were cultured in high-glucose DMEM (GIBCO, USA) media supplemented with 10% fetal bovine serum (FBS) and 1% penicillin–streptomycin, and incubated in a humidified environment at 37 °C with 5% CO2. Oxyhemoglobin (10 μM, Shanghai Yuanye Bio-Technology Co., China) was added to the medium for 24 h to induce in vitro SAH [25]. For the control +IL-4 group and oxyhemoglobin + IL-4 group, 20 ng/ml IL-4 was administered for 24 h of treatment with reference to previous descriptions before [26] proceeding to the next step of the study. DMSO was used as a control for the treatment conditions.

Phagocytosis Assay

As described above, BV2 cells were inoculated into 24-well plates and incubated for 24 h, and then the in vitro SAH model was induced, followed by 24 h of incubation with or without IL-4. Fluorescent microbeads with a diameter of 1 μm (Invitrogen, diluted 1:15,000) were introduced into the culture medium and incubated with the cells for 4 h. The BV2 cells were then fixed on cover slips using 4% paraformaldehyde. Immunofluorescent staining with phalloidin was performed to label the cell cytoskeleton, while DAPI was used to stain the cell nuclei for 5 min. Laser confocal microscopy was used for imaging, and Image J software was used to count the number of microglia phagocytosed microspheres in different groups by randomly selecting 3 fields of view within 4 wells in each group [27, 28].

qRT-PCR

Total RNA was extracted from rat brain basal cortex tissue and BV2 cells using the Total RNA Kit (Qiagen, Santa Clara, CA). The isolated RNA was then reverse transcribed into complementary DNA (cDNA) through the reverse transcription process, employing the First Strand cDNA Synthesis Kit (Yeasen, Shanghai, China) according to the manufacturer’s instructions. Amplification was subsequently performed as per the manufacturer’s protocol. The endogenous reference gene primers for GAPDH in rats and mice, as well as other qRT-PCR primers, were obtained from Sangon Technology (Shanghai, China) and are listed in Table 1. All experiments were repeated three times. The relative expression levels were calculated using the 2 − ΔΔCt method, with GAPDH mRNA serving as an internal control for normalization.
Table 1
Primers Used in the qRT-PCR Reaction
 
Gene
Forward primer (5′-3′)
Reverse primer (5′-3′)
For rat experiments
Tnf α
CCCAGACCCTCACACTCAGATCAT
CAGCCTTGTCCCTTGAAGAGAA
iNOS
CAAGCACCTTGGAAGAGGAG
AAGGCCAAACACAGCATACC
CD32
AATCCTGCCGTTCCTACTGATC
GTGTCACCGTGTCTTCCTTGAG
IL-1β
TCTCACAGCAGCATCTCGACAAG
CCACGGGCAAGACATAGGTAGC
IL-10
AAGGCAGTGGAGCAGGTGAAG
CACGTAGGCTTCTATGCAGTTGATG
CCL-22
CTGATGCAGGTCCCTATGGT
GCAGGATTTTGAGGTCCAGA
For BV2 cell experiments
iNOS
CAAGCACCTTGGAAGAGGAG
AAGGCCAAACACAGCATACC
Arg1
TCACCTGAGCTTTGATGTCG
CTGAAAGGAGCCCTGTCTTG
Aqp1
TTGACTACACTGGCTGCGGTATC
GTTTGAGAAGTTGCGGGTGAGC
Mgl2
CTAACAGTTCCTTCCCAGTCCTTCC
CACGGAGATGACCACCAGTAGC
Mmp13
CTTCCTGATGATGACGTTCAAG
GTCACACTTCTCTGGTGTTTTG
Cybb
GACAGGAACCTCACTTTCCATA
TGAAGAGATGTGCAATTGTGTG
Mmp12
TGTACAGCATCTTAGAGCAGTG
TATGTAGTCTACATCCTCACGC
Cx3cr1
TCGGTCTGGTGGGAAATCTGTTG
CAGGTTCAGGAGGTAGATGTCAGTG
Ccl9
CTGCCCTCTCCTTCCTCATTCTTAC
TGCTGTGCCTTCAGACTGCTC

RNA Sequencing (RNA-Seq) Analysis

Total RNA was isolated from BV2 cells of both the oxyhemoglobin group and oxyhemoglobin + IL-4 group, with subsequent sequencing analysis primarily carried out by BGI Corporation (China) [29]. Differential expression analysis was carried out using DESeq2 with a log-fold change (FC) threshold of > 0.5 and q value (p-adjusted) < 0.05 to identify differentially expressed genes (DEGs). Gene ontology (GO) analysis, KEGG pathway analysis, and protein–protein interaction (PPI) network analysis were all performed on the Dr. Tom network platform of BGI (online analysis and visualization website: http://​report.​bgi.​com), and q value < 0.05 was considered pathway enrichment. The protein–protein interaction (PPI) was mapped by String (https://​string-db.​org/​).

Statistical Analysis of Data

All data in this study were presented as mean ± standard deviation (SD) and analyzed using GraphPad Prism 9.0 software (USA). Unpaired Student t-test was used to compare the two groups, while one-way analysis of variance (ANOVA) was used between multiple groups followed by Tukey’s post hoc tests. The Mann–Whitney test (for two groups) or the Kruskal–Wallis test followed by Dunn’s post hoc test (for multiple groups) was employed to examine variables that did not conform to a normal distribution. Two-way ANOVA with Bonferroni post hoc test was performed for modified Garcia score and hang wire test. p < 0.05 was considered statistically significant.

RESULTS

The Expression Level of IL-4 in CSF Was Increased After SAH

In this study, we measured the expression levels of IL-4 in peripheral blood serum and CSF of rats at 12 h, 24 h, 3 days, and 5days after SAH using ELISA. The results showed that compared to the sham group rats, there was no significant change in IL-4 expression levels in the serum at all time points after SAH (Fig. 1b, p > 0.05). However, we observed a significant increase in IL-4 expression in the CSF of rats at 12 h and 5 days after SAH (Fig. 1c, p < 0.05).

IL-4 Administration Improves Neurological Deficits Following SAH

Furthermore, we examined the impact of IL-4 administration on neurological deficits after SAH. The endovascular perforation model was utilized to induce SAH in rats, and on the 5th day post-surgery, visible blood clots were observed in the Willis circle of all SAH groups, with no statistically significant differences in SAH grading scores among the different SAH groups (Fig. 1d–e, p > 0.05). To assess the potential improvement in sensory-motor function after SAH, modified Garcia tests and hang wire tests were performed at 1, 3, and 5 days post-SAH. The results showed that there was no statistically significant difference in neurological function scores between the sham group and the sham + IL-4 group at each observed time point (p > 0.05). Compared to the sham group, the SAH group exhibited significant neurological functional impairments, which gradually recovered over time. However, treatment with IL-4 facilitated this functional recovery, as indicated by the modified Garcia test and hang wire test scores at 5 days (Fig. 1f, g, p < 0.05). These findings suggest that IL-4 administration can improve neurological deficits after SAH.

IL-4 Administration Attenuates Neuronal Apoptosis After SAH

Subsequently, we assessed the potential of continuous ventricular delivery of IL-4 over a 5-day period to rescue neuronal damage following SAH. Immunofluorescent staining targeting the apoptosis-related factor cleaved caspase-3 and the neuronal marker NeuN was performed in the basal cortex of the brain post-SAH (Fig. 2). The results depicted in Fig. 2b demonstrate a significant increase in the number of cleaved caspase-3-positive neurons in the basal cortex of the SAH group compared to the sham group after 5 days, whereas IL-4 treatment significantly decreased this number (p < 0.05). There was no significant difference between sham + IL-4 group and sham group (p > 0.05). Furthermore, the immunofluorescence intensity of cleaved caspase-3 was quantified, revealing a marked reduction in its levels in the basal cortex after SAH with IL-4 treatment (Fig. 2c, p < 0.05). These findings provide evidence that IL-4 administration can ameliorate cellular apoptosis in rats induced with SAH.

IL-4 Promotes Microglial Phenotype Switch and Alleviates Neuroinflammation After SAH

To investigate the effects of IL-4 on microglial polarization after SAH, we conducted double immunostaining of Iba1 with M1 phenotype marker CD68 and M2 phenotype marker CD206 in the basal cortex. Our results demonstrated a significant increase in the number of CD68-positive Iba1 cells, indicating an activation of M1 microglia, after SAH compared to the sham group. However, treatment with IL-4 significantly reduced the co-localization of CD68-positive Iba1 cells (Fig. 3a, c, p < 0.05). Furthermore, we examined the expression of M2 microglia and found a significant increase in CD206-positive Iba1 cells after IL-4 treatment compared to the SAH group (Fig. 3b, d, p < 0.05). Notably, IL-4 treatment did not alter the activation of microglia in sham rats. These findings suggest that IL-4 has a positive impact on microglial phenotype switch and can alleviate neuroinflammation after SAH.
We conducted ELISA to assess the expression levels of inflammatory factors TNF-α, iNOS, and IL-10 in the ipsilateral basal cortex of rat brain tissue after SAH (Fig. 4a). The results showed that IL-4 treatment did not significantly alter the expression levels of TNF-α, iNOS, and IL-10 in the sham group (p > 0.5). However, SAH induction increased the expression levels of TNF-α, iNOS, and IL-10. Notably, IL-4 treatment significantly reduced the expression levels of pro-inflammatory cytokines TNF-α and iNOS, while simultaneously increasing the level of the anti-inflammatory cytokine IL-10.
We further validated the expression of six cytokines associated with neuroinflammation in the sham group, SAH group, and SAH + IL-4 group using qRT-PCR. Five days after SAH, compared to the sham group, the expression of pro-inflammatory markers (including TNF-α, iNOS, CD32, IL-1β) in the brains of SAH rats treated with the vehicle was significantly increased. However, after 5 days of IL-4 treatment, the expression of TNF-α, iNOS, and IL-1β was significantly suppressed. Additionally, the anti-inflammatory cytokine IL-10 was observed to be upregulated following the occurrence of SAH, and both CCL-22 and IL-10 exhibited a substantial increase after a 5-day administration of IL-4 (Fig. 4b, p < 0.05). These findings suggest that IL-4 treatment possesses the capacity to regulate the neuroinflammatory response subsequent to SAH.

IL-4 Enhances the Phagocytic Efficiency of Microglial Cells and Regulates the Inflammatory Response In Vitro

The engulfment of cellular debris and deceased or damaged cells represents a pivotal function of microglial cells in the context of inflammatory immune reactions. To gain deeper insights into the association between IL-4 administration and microglial cell regulation, we induced the SAH model in vitro and administered IL-4 in the treatment group. Through quantitative analysis utilizing fluorescent microbeads, it was observed that cellular phagocytosis was significantly enhanced in the SAH model in vitro (p < 0.001), and the phagocytic functionality of the oxyhemoglobin + IL-4 group significantly surpassed that of the oxyhemoglobin group (Fig. 5a–b, p < 0.05). IL-4 had no effect on the phagocytosis of BV-2 cells in the control group.
We next assessed the role of IL-4 treatment in inflammatory response in BV2 microglial cells using qRT-PCR and ELISA. The results from ELISA analysis indicated that there was no significant difference in the expression levels of TNF-α, iNOS, and IL-10 between the control + IL-4 group and the control group (Fig. 5e). In the in vitro SAH model, we observed a substantial increase in TNF-α and iNOS levels in BV2 cells compared to the control group. However, when IL-4 treatment was administered, the levels of TNF-α and iNOS were significantly reduced. Additionally, IL-4 treatment resulted in a significant increase in the level of the anti-inflammatory cytokine IL-10 after SAH (p < 0.05). The results of qRT-PCR were largely consistent with the results of the in vivo experiments, in which the in vitro SAH model resulted in heightened levels of the M1 pro-inflammatory marker iNOS in microglial cells, whereas the levels of iNOS were significantly reduced after IL-4 administration (Fig. 5c). Concurrently, IL-4 treatment significantly resulted in a significant elevation in the expression levels of the M2 phenotype pro-inflammatory marker Arg-1 (Fig. 5d, p < 0.05). These results underline the regulatory role of IL-4 on the phagocytic function and inflammatory response of microglial cells in vitro.

RNA-Seq Analysis of BV2 Cells In Vitro

To elucidate the biological processes that may be altered by IL-4 interference, we conducted RNA-Seq analysis on BV2 microglial cells in oxyhemoglobin and oxyhemoglobin + IL-4 groups. We identified a total of 72 differentially expressed genes (DEGs), consisting of 21 upregulated genes and 51 downregulated genes (Fig. 6a), of which 10 upregulated and 10 downregulated genes were depicted in the heatmap (Fig. 6b). Subsequently, we conducted Gene Ontology (GO) enrichment analysis on these 72 commonly regulated DEGs. GO enrichment unveiled significant statistical enrichment of genes involved in diverse biological processes, encompassing immune response, immune system process, inflammatory response, positive regulation of gene expression, and positive regulation of ERK1 and ERK2 cascade (Fig. 6c). In terms of cellular component categorization, the top-enriched cluster encompassed plasma membrane, cell surface, and the external side of the plasma membrane (Fig. 6d). Functional analysis of DEGs enrichment primarily associated with cytokine receptor activity, CCR1 chemokine receptor binding, and CD4 receptor binding (Fig. 6e).
Subsequently, we performed KEGG pathway enrichment and PPI analysis of the DEGs. The highest ranked pathway based on the number of genes involved was the cytokine-cytokine receptor interaction pathway (Fig. 7a). In the PPI network, the 10 key genes regulating IL-4 in SAH-induced neuroinflammation were Tnf, Tlr2, Arg1, Ccl3, Cxcr2, Cx3cr1, Cybb, Hp, Cd74, and Ccl9, with Tnf being associated with a variety of biological processes (Fig. 7b). Hence, we hypothesize that Tnf plays a crucial role in the IL-4-mediated modulation of neuroinflammation following SAH. The relative levels of upregulated genes Aqp1, Mg12, Mmp13, Mmp12, and Ccl9 and downregulated genes Cybb and Cx3cr1 were verified by qRT-PCR (Fig. 7c).

DISCUSSION

IL-4 is a mammalian-specific cytokine that plays a pivotal role as a pleiotropic cytokine in numerous immune/inflammatory pathways [14], particularly in the development of type 2 inflammatory responses and the polarization of macrophages towards an M2 phenotype [30, 31]. As a significant anti-inflammatory, it plays a protective role in brain tissue after ischemic injury [22]. However, its role in the pathophysiology of SAH remains unclear.
Clinical studies in SAH patients have reported elevated [15] or unchanged levels [16] of IL-4 in peripheral blood during the acute phase, whereas the levels of IL-4 in the CSF are elevated and have been considered a protective factor associated with survival [17] and early compensatory anti-inflammatory response [16]. In experimental animal models of SAH, activation of A3 adenosine receptors upregulated IL-4 expression and ameliorated early brain injury following SAH [22]. Therefore, enhancing IL-4 levels holds promising therapeutic potential in promoting neurofunctional recovery after SAH. In our study, we observed no significant change in serum IL-4 expression levels in rats after SAH, while expression levels in the CSF significantly increased at 12 h and 5 days. This increase is believed to be, at least in part, a result of the infiltration and accumulation of T cells and other immune cells in the injured brain.
Neuroinflammation is a major pathological hallmark following SAH. Activation of resident microglia in the CNS and infiltration of macrophages are key events targeted by IL-4 for SAH treatment [20]. While it is well-established that IL-4 acts as a promoter of M2 macrophage polarization [32], the impact of exogenous IL-4 on the polarization state of microglia/macrophages after SAH has not been previously investigated. Here, we explore the functional response of BV2 microglial cells in SAH animal models and in vitro culture upon IL-4 administration. In the animal models, IL-4 administration resulted in neurological functional recovery, reduced apoptosis of cortical neurons, phenotypic transition of microglia towards an anti-inflammatory M2 phenotype, and suppression of pro-inflammatory factors including TNF-α, iNOS, and IL-1β. In vitro experiments demonstrated that IL-4 enhanced the phagocytic capacity of microglial cells and alleviated neuroinflammation. In the current study, we found that IL-4 treatment did not have an impact on microglia activation in sham rats or the regulation of inflammation and phagocytosis in control BV2 cells, which is consistent with the results of a recent study [33]. It is worth noting that previous studies have mainly focused on the long-term neuroprotective effects of IL-4 [22, 34], whereas our results demonstrate its beneficial effects on neurofunction occurring at 5 days post-SAH. Although longer-term observation was not carried out, our findings suggest significant potential for sustained functional recovery.
RNA-Seq analysis of microglia confirmed that IL-4 regulates various inflammatory pathways, primarily through cytokine-cytokine receptor interaction. Tnf, Tlr2, Arg1, Ccl3, Cxcr2, Cx3cr1, Cybb, Hp, Cd74, and Ccl9 were identified as key driver genes regulated by IL-4 administration in vitro SAH models. Among them, Tnf is associated with multiple biological processes including cytokine-cytokine receptor interaction and cell apoptosis. The TNF-α signaling pathway is widely recognized to assume a crucial role in the pathogenesis of SAH, and its signaling process is intricate, regulating SAH through many interconnected pathways. Targeting TNF-α function has the potential to ameliorate SAH-induced brain damage and serve as a therapeutic target [3537].
It is noteworthy that we discovered an upregulation of the chemokine receptor CXCR2 in microglial cells after IL-4 treatment. A study examining immune-regulatory biomarkers in blood and tissue samples of SAH patients also indicates the involvement of CXCR2 in SAH progression [38]. While it is known that CXCR2 is upregulated after stroke to recruit neutrophils to the brain, blocking CXCR2 does not necessarily improve neurofunctional outcomes [3941]. Rapid downregulation of CXCR2 may impact neuronal-glial communication [42]. Some researchers have also found that IL-4 incubation upregulates CXCR2 expression in human monocytes and macrophages [43], and the combination of IL-4 and TNF-α induces autocrine secretion of CXCR2 chemokines [44]. However, further investigation is required to clarify the impact of IL-4 on CXCR2 after SAH and its contribution to SAH. Overall, current research suggests that the beneficial effects of IL-4 cannot be attributed solely to the actions of a single cell type but rather to complex cascade reactions.
Despite promising results of IL-4 treatment in animal models of CNS diseases, further research is imperative to determine the most optimal delivery route of administration for patients. Intravenous delivery holds prominence in clinical practice; however, it is essential to note that IL-4 exhibits a markedly abbreviated half-life [45, 46]. In the context of targeting cerebrovascular accidents, as exemplified in our investigation, the administration of IL-4 into the CSF is frequently employed. Nevertheless, it is imperative to acknowledge that the employment of stereotactic surgical techniques in our research is invasive and engenders potential hazards, including infection and surgery-induced secondary brain damage. Consequently, the generalizability of this approach to subarachnoid hemorrhage (SAH) patients may be restricted. Gene engineering approaches have been explored for IL-4 expression [47], but the optimal clinical delivery method remains to be investigated. In addition, the BV2 microglial cell model in vitro has helped to elucidate the molecular mechanisms of IL-4 therapy but has not been studied on neurons or astrocytes, which in fact communicate with each other and play multiple roles in the presence of IL-4 [48, 49].
In conclusion, this study demonstrates, for the first time, that exogenous IL-4 administration effectively enhances neurobehavioral performance and mitigates neuronal apoptosis post-SAH, which may be attributed to a protective mechanism that induces M2 phenotypic transformation in microglia/macrophages. IL-4 governs the polarization of microglia and the secretion of pro-inflammatory cytokines by means of complex signaling pathways, including cytokine-cytokine receptor interactions.

Declarations

Ethics Approval

All animal experiments were approved by the Institutional Animal Care and Use Committee of Shandong First Medical University and conducted in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals.

Competing Interests

The authors declare no competing interests.
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/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Dent – Das Online-Abo der Zahnmedizin

Online-Abonnement

Mit e.Dent erhalten Sie Zugang zu allen zahnmedizinischen Fortbildungen und unseren zahnmedizinischen und ausgesuchten medizinischen Zeitschriften.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Etminan, N., H.S. Chang, K. Hackenberg, N.K. de Rooij, M.D.I. Vergouwen, G.J.E. Rinkel, and A. Algra. 2019. Worldwide incidence of aneurysmal subarachnoid hemorrhage according to region, time period, blood pressure, and smoking prevalence in the population: A systematic review and meta-analysis. JAMA Neurology 76: 588–597.PubMedPubMedCentralCrossRef Etminan, N., H.S. Chang, K. Hackenberg, N.K. de Rooij, M.D.I. Vergouwen, G.J.E. Rinkel, and A. Algra. 2019. Worldwide incidence of aneurysmal subarachnoid hemorrhage according to region, time period, blood pressure, and smoking prevalence in the population: A systematic review and meta-analysis. JAMA Neurology 76: 588–597.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Taufique, Z., T. May, E. Meyers, C. Falo, S.A. Mayer, S. Agarwal, S. Park, E.S. Connolly, J. Claassen, and J.M. Schmidt. 2016. Predictors of poor quality of life 1 year after subarachnoid hemorrhage. Neurosurgery 78: 256–264.PubMedCrossRef Taufique, Z., T. May, E. Meyers, C. Falo, S.A. Mayer, S. Agarwal, S. Park, E.S. Connolly, J. Claassen, and J.M. Schmidt. 2016. Predictors of poor quality of life 1 year after subarachnoid hemorrhage. Neurosurgery 78: 256–264.PubMedCrossRef
3.
Zurück zum Zitat Lauzier, D.C., K. Jayaraman, J.Y. Yuan, D. Diwan, A.K. Vellimana, J.W. Osbun, A.R. Chatterjee, U. Athiraman, R. Dhar, and G.J. Zipfel. 2023. Early brain injury after subarachnoid hemorrhage: Incidence and mechanisms. Stroke 54: 1426–1440.PubMedCrossRef Lauzier, D.C., K. Jayaraman, J.Y. Yuan, D. Diwan, A.K. Vellimana, J.W. Osbun, A.R. Chatterjee, U. Athiraman, R. Dhar, and G.J. Zipfel. 2023. Early brain injury after subarachnoid hemorrhage: Incidence and mechanisms. Stroke 54: 1426–1440.PubMedCrossRef
4.
Zurück zum Zitat Sehba, F.A., J. Hou, R.M. Pluta, and J.H. Zhang. 2012. The importance of early brain injury after subarachnoid hemorrhage. Progress in Neurobiology 97: 14–37.PubMedPubMedCentralCrossRef Sehba, F.A., J. Hou, R.M. Pluta, and J.H. Zhang. 2012. The importance of early brain injury after subarachnoid hemorrhage. Progress in Neurobiology 97: 14–37.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Schneider, U.C., A.M. Davids, S. Brandenburg, A. Müller, A. Elke, S. Magrini, E. Atangana, K. Turkowski, T. Finger, A. Gutenberg, et al. 2015. Microglia inflict delayed brain injury after subarachnoid hemorrhage. Acta Neuropathologica 130: 215–231.PubMedCrossRef Schneider, U.C., A.M. Davids, S. Brandenburg, A. Müller, A. Elke, S. Magrini, E. Atangana, K. Turkowski, T. Finger, A. Gutenberg, et al. 2015. Microglia inflict delayed brain injury after subarachnoid hemorrhage. Acta Neuropathologica 130: 215–231.PubMedCrossRef
6.
Zurück zum Zitat Schneider, U.C., R. Xu, and P. Vajkoczy. 2018. Inflammatory events following subarachnoid hemorrhage (SAH). Current Neuropharmacology 16: 1385–1395.PubMedPubMedCentralCrossRef Schneider, U.C., R. Xu, and P. Vajkoczy. 2018. Inflammatory events following subarachnoid hemorrhage (SAH). Current Neuropharmacology 16: 1385–1395.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Kigerl, K.A., J.C. Gensel, D.P. Ankeny, J.K. Alexander, D.J. Donnelly, and P.G. Popovich. 2009. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. Journal of Neuroscience 29: 13435–13444.PubMedCrossRef Kigerl, K.A., J.C. Gensel, D.P. Ankeny, J.K. Alexander, D.J. Donnelly, and P.G. Popovich. 2009. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. Journal of Neuroscience 29: 13435–13444.PubMedCrossRef
8.
Zurück zum Zitat Ansari, M.A. 2015. Temporal profile of M1 and M2 responses in the hippocampus following early 24h of neurotrauma. Journal of the Neurological Sciences 357: 41–49.PubMedCrossRef Ansari, M.A. 2015. Temporal profile of M1 and M2 responses in the hippocampus following early 24h of neurotrauma. Journal of the Neurological Sciences 357: 41–49.PubMedCrossRef
9.
Zurück zum Zitat Orihuela, R., C.A. McPherson, and G.J. Harry. 2016. Microglial M1/M2 polarization and metabolic states. British Journal of Pharmacology 173: 649–665.PubMedCrossRef Orihuela, R., C.A. McPherson, and G.J. Harry. 2016. Microglial M1/M2 polarization and metabolic states. British Journal of Pharmacology 173: 649–665.PubMedCrossRef
10.
Zurück zum Zitat Zhang, J., P. Rong, L. Zhang, H. He, T. Zhou, Y. Fan, L. Mo, Q. Zhao, Y. Han, S. Li, et al. 2021. IL4-driven microglia modulate stress resilience through BDNF-dependent neurogenesis. Science Advances 7. Zhang, J., P. Rong, L. Zhang, H. He, T. Zhou, Y. Fan, L. Mo, Q. Zhao, Y. Han, S. Li, et al. 2021. IL4-driven microglia modulate stress resilience through BDNF-dependent neurogenesis. Science Advances 7.
11.
Zurück zum Zitat Hasan, M., J.E. Seo, K.A. Rahaman, M.J. Kang, B.H. Jung, and O.S. Kwon. 2016. Increased levels of brain serotonin correlated with MMP-9 activity and IL-4 levels resulted in severe experimental autoimmune encephalomyelitis (EAE) in obese mice. Neuroscience 319: 168–182.PubMedCrossRef Hasan, M., J.E. Seo, K.A. Rahaman, M.J. Kang, B.H. Jung, and O.S. Kwon. 2016. Increased levels of brain serotonin correlated with MMP-9 activity and IL-4 levels resulted in severe experimental autoimmune encephalomyelitis (EAE) in obese mice. Neuroscience 319: 168–182.PubMedCrossRef
12.
Zurück zum Zitat Zhao, X., H. Wang, G. Sun, J. Zhang, N.J. Edwards, and J. Aronowski. 2015. Neuronal interleukin-4 as a modulator of microglial pathways and ischemic brain damage. Journal of Neuroscience 35: 11281–11291.PubMedCrossRef Zhao, X., H. Wang, G. Sun, J. Zhang, N.J. Edwards, and J. Aronowski. 2015. Neuronal interleukin-4 as a modulator of microglial pathways and ischemic brain damage. Journal of Neuroscience 35: 11281–11291.PubMedCrossRef
13.
Zurück zum Zitat Ponomarev, E.D., K. Maresz, Y. Tan, and B.N. Dittel. 2007. CNS-derived interleukin-4 is essential for the regulation of autoimmune inflammation and induces a state of alternative activation in microglial cells. Journal of Neuroscience 27: 10714–10721.PubMedCrossRef Ponomarev, E.D., K. Maresz, Y. Tan, and B.N. Dittel. 2007. CNS-derived interleukin-4 is essential for the regulation of autoimmune inflammation and induces a state of alternative activation in microglial cells. Journal of Neuroscience 27: 10714–10721.PubMedCrossRef
14.
Zurück zum Zitat Gärtner, Y., L. Bitar, F. Zipp, and C.F. Vogelaar. 2023. Interleukin-4 as a therapeutic target. Pharmacology & Therapeutics 242: 108348.CrossRef Gärtner, Y., L. Bitar, F. Zipp, and C.F. Vogelaar. 2023. Interleukin-4 as a therapeutic target. Pharmacology & Therapeutics 242: 108348.CrossRef
15.
Zurück zum Zitat Zhou, Y., Y. Jiang, Y. Peng, and M. Zhang. 2017. The quantitative and functional changes of postoperative peripheral blood immune cell subsets relate to prognosis of patients with subarachnoid hemorrhage: A preliminary study. World Neurosurgery 108: 206–215.PubMedCrossRef Zhou, Y., Y. Jiang, Y. Peng, and M. Zhang. 2017. The quantitative and functional changes of postoperative peripheral blood immune cell subsets relate to prognosis of patients with subarachnoid hemorrhage: A preliminary study. World Neurosurgery 108: 206–215.PubMedCrossRef
16.
Zurück zum Zitat Al-Tamimi, Y.Z., D. Bhargava, N.M. Orsi, A. Teraifi, M. Cummings, U.V. Ekbote, A.C. Quinn, S. Homer-Vanniasinkam, and S. Ross. 2019. Compartmentalisation of the inflammatory response following aneurysmal subarachnoid haemorrhage. Cytokine 123: 154778.PubMedCrossRef Al-Tamimi, Y.Z., D. Bhargava, N.M. Orsi, A. Teraifi, M. Cummings, U.V. Ekbote, A.C. Quinn, S. Homer-Vanniasinkam, and S. Ross. 2019. Compartmentalisation of the inflammatory response following aneurysmal subarachnoid haemorrhage. Cytokine 123: 154778.PubMedCrossRef
17.
Zurück zum Zitat Righy, C., R. Turon, G. Freitas, A.M. Japiassú, H.C.C. Faria Neto, M. Bozza, M.F. Oliveira, and F.A. Bozza. 2018. Hemoglobin metabolism by-products are associated with an inflammatory response in patients with hemorrhagic stroke. Revista Brasileira de Terapia Intensiva 30: 21–27.PubMedPubMedCentralCrossRef Righy, C., R. Turon, G. Freitas, A.M. Japiassú, H.C.C. Faria Neto, M. Bozza, M.F. Oliveira, and F.A. Bozza. 2018. Hemoglobin metabolism by-products are associated with an inflammatory response in patients with hemorrhagic stroke. Revista Brasileira de Terapia Intensiva 30: 21–27.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Sugawara, T., R. Ayer, V. Jadhav, and J.H. Zhang. 2008. A new grading system evaluating bleeding scale in filament perforation subarachnoid hemorrhage rat model. Journal of Neuroscience Methods 167: 327–334.PubMedCrossRef Sugawara, T., R. Ayer, V. Jadhav, and J.H. Zhang. 2008. A new grading system evaluating bleeding scale in filament perforation subarachnoid hemorrhage rat model. Journal of Neuroscience Methods 167: 327–334.PubMedCrossRef
19.
Zurück zum Zitat Chen, Q., Y. Cai, X. Zhu, J. Wang, F. Gao, M. Yang, L. Mao, Z. Zhang, and B. Sun. 2022. Edaravone dexborneol treatment attenuates neuronal apoptosis and improves neurological function by suppressing 4-HNE-associated oxidative stress after subarachnoid hemorrhage. Frontiers in Pharmacology 13: 848529.PubMedPubMedCentralCrossRef Chen, Q., Y. Cai, X. Zhu, J. Wang, F. Gao, M. Yang, L. Mao, Z. Zhang, and B. Sun. 2022. Edaravone dexborneol treatment attenuates neuronal apoptosis and improves neurological function by suppressing 4-HNE-associated oxidative stress after subarachnoid hemorrhage. Frontiers in Pharmacology 13: 848529.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Mao, L., L. Sun, J. Sun, B. Sun, Y. Gao, and H. Shi. 2021. Ethyl pyruvate improves white matter remodeling in rats after traumatic brain injury. CNS Neuroscience & Therapeutics 27: 113–122.CrossRef Mao, L., L. Sun, J. Sun, B. Sun, Y. Gao, and H. Shi. 2021. Ethyl pyruvate improves white matter remodeling in rats after traumatic brain injury. CNS Neuroscience & Therapeutics 27: 113–122.CrossRef
21.
Zurück zum Zitat Toft-Bertelsen, T.L., D. Barbuskaite, E.K. Heerfordt, S.D. Lolansen, S.N. Andreassen, N. Rostgaard, M.H. Olsen, N.H. Norager, T. Capion, M.F. Rath, et al. 2022. Lysophosphatidic acid as a CSF lipid in posthemorrhagic hydrocephalus that drives CSF accumulation via TRPV4-induced hyperactivation of NKCC1. Fluids Barriers CNS 19: 69.PubMedPubMedCentralCrossRef Toft-Bertelsen, T.L., D. Barbuskaite, E.K. Heerfordt, S.D. Lolansen, S.N. Andreassen, N. Rostgaard, M.H. Olsen, N.H. Norager, T. Capion, M.F. Rath, et al. 2022. Lysophosphatidic acid as a CSF lipid in posthemorrhagic hydrocephalus that drives CSF accumulation via TRPV4-induced hyperactivation of NKCC1. Fluids Barriers CNS 19: 69.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Liu, X., J. Liu, S. Zhao, H. Zhang, W. Cai, M. Cai, X. Ji, R.K. Leak, Y. Gao, J. Chen, and X. Hu. 2016. Interleukin-4 is essential for microglia/macrophage M2 polarization and long-term recovery after cerebral ischemia. Stroke 47: 498–504.PubMedPubMedCentralCrossRef Liu, X., J. Liu, S. Zhao, H. Zhang, W. Cai, M. Cai, X. Ji, R.K. Leak, Y. Gao, J. Chen, and X. Hu. 2016. Interleukin-4 is essential for microglia/macrophage M2 polarization and long-term recovery after cerebral ischemia. Stroke 47: 498–504.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Wang, J., Y. Cai, J. Sun, H. Feng, X. Zhu, Q. Chen, F. Gao, Q. Ni, L. Mao, M. Yang, and B. Sun. 2023. Administration of intramuscular AAV-BDNF and intranasal AAV-TrkB promotes neurological recovery via enhancing corticospinal synaptic connections in stroke rats. Experimental Neurology 359: 114236.PubMedCrossRef Wang, J., Y. Cai, J. Sun, H. Feng, X. Zhu, Q. Chen, F. Gao, Q. Ni, L. Mao, M. Yang, and B. Sun. 2023. Administration of intramuscular AAV-BDNF and intranasal AAV-TrkB promotes neurological recovery via enhancing corticospinal synaptic connections in stroke rats. Experimental Neurology 359: 114236.PubMedCrossRef
24.
Zurück zum Zitat Hu, Q., Q. Du, W. Yu, and X. Dong. 2022. 2-Methoxyestradiol alleviates neuroinflammation and brain edema in early brain injury after subarachnoid hemorrhage in rats. Frontiers in Cellular Neuroscience 16: 869546.PubMedPubMedCentralCrossRef Hu, Q., Q. Du, W. Yu, and X. Dong. 2022. 2-Methoxyestradiol alleviates neuroinflammation and brain edema in early brain injury after subarachnoid hemorrhage in rats. Frontiers in Cellular Neuroscience 16: 869546.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Sun, X.G., X.H. Chu, I.S. Godje Godje, S.Y. Liu, H.Y. Hu, Y.B. Zhang, L.J. Zhu, H. Wang, C. Sui, J. Huang, and Y.J. Shen. 2022. Aerobic glycolysis induced by mTOR/HIF-1α promotes early brain injury after subarachnoid hemorrhage via activating M1 microglia. Translational Stroke Research. Sun, X.G., X.H. Chu, I.S. Godje Godje, S.Y. Liu, H.Y. Hu, Y.B. Zhang, L.J. Zhu, H. Wang, C. Sui, J. Huang, and Y.J. Shen. 2022. Aerobic glycolysis induced by mTOR/HIF-1α promotes early brain injury after subarachnoid hemorrhage via activating M1 microglia. Translational Stroke Research.
26.
Zurück zum Zitat Latta, C.H., T.L. Sudduth, E.M. Weekman, H.M. Brothers, E.L. Abner, G.J. Popa, M.D. Mendenhall, F. Gonzalez-Oregon, K. Braun, and D.M. Wilcock. 2015. Determining the role of IL-4 induced neuroinflammation in microglial activity and amyloid-β using BV2 microglial cells and APP/PS1 transgenic mice. Journal of Neuroinflammation 12: 41.PubMedPubMedCentralCrossRef Latta, C.H., T.L. Sudduth, E.M. Weekman, H.M. Brothers, E.L. Abner, G.J. Popa, M.D. Mendenhall, F. Gonzalez-Oregon, K. Braun, and D.M. Wilcock. 2015. Determining the role of IL-4 induced neuroinflammation in microglial activity and amyloid-β using BV2 microglial cells and APP/PS1 transgenic mice. Journal of Neuroinflammation 12: 41.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Nakamura, R., M. Konishi, Y. Higashi, M. Saito, and T. Akizawa. 2023. Five-mer peptides prevent short-term spatial memory deficits in Aβ25-35-induced Alzheimer’s model mouse by suppressing Aβ25-35 aggregation and resolving its aggregate form. Alzheimer’s Research & Therapy 15: 83.CrossRef Nakamura, R., M. Konishi, Y. Higashi, M. Saito, and T. Akizawa. 2023. Five-mer peptides prevent short-term spatial memory deficits in Aβ25-35-induced Alzheimer’s model mouse by suppressing Aβ25-35 aggregation and resolving its aggregate form. Alzheimer’s Research & Therapy 15: 83.CrossRef
28.
Zurück zum Zitat Cui, W., C. Sun, Y. Ma, S. Wang, X. Wang, and Y. Zhang. 2020. Inhibition of TLR4 induces M2 microglial polarization and provides neuroprotection via the NLRP3 inflammasome in Alzheimer’s disease. Frontiers in Neuroscience 14: 444.PubMedPubMedCentralCrossRef Cui, W., C. Sun, Y. Ma, S. Wang, X. Wang, and Y. Zhang. 2020. Inhibition of TLR4 induces M2 microglial polarization and provides neuroprotection via the NLRP3 inflammasome in Alzheimer’s disease. Frontiers in Neuroscience 14: 444.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Ma, Z., D. Liu, W. Li, S. Di, Z. Zhang, J. Zhang, L. Xu, K. Guo, Y. Zhu, J. Han, et al. 2019. STYK1 promotes tumor growth and metastasis by reducing SPINT2/HAI-2 expression in non-small cell lung cancer. Cell Death & Disease 10: 435.CrossRef Ma, Z., D. Liu, W. Li, S. Di, Z. Zhang, J. Zhang, L. Xu, K. Guo, Y. Zhu, J. Han, et al. 2019. STYK1 promotes tumor growth and metastasis by reducing SPINT2/HAI-2 expression in non-small cell lung cancer. Cell Death & Disease 10: 435.CrossRef
30.
Zurück zum Zitat Bernstein, Z.J., A. Shenoy, A. Chen, N.M. Heller, and J.B. Spangler. 2023. Engineering the IL-4/IL-13 axis for targeted immune modulation. Immunological Reviews. Bernstein, Z.J., A. Shenoy, A. Chen, N.M. Heller, and J.B. Spangler. 2023. Engineering the IL-4/IL-13 axis for targeted immune modulation. Immunological Reviews.
31.
Zurück zum Zitat Czimmerer, Z., B. Daniel, A. Horvath, D. Rückerl, G. Nagy, M. Kiss, M. Peloquin, M.M. Budai, I. Cuaranta-Monroy, Z. Simandi, et al. 2018. The transcription factor STAT6 mediates direct repression of inflammatory enhancers and limits activation of alternatively polarized macrophages. Immunity 48: 75-90.e76.PubMedPubMedCentralCrossRef Czimmerer, Z., B. Daniel, A. Horvath, D. Rückerl, G. Nagy, M. Kiss, M. Peloquin, M.M. Budai, I. Cuaranta-Monroy, Z. Simandi, et al. 2018. The transcription factor STAT6 mediates direct repression of inflammatory enhancers and limits activation of alternatively polarized macrophages. Immunity 48: 75-90.e76.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Kawahara, K., M. Suenobu, A. Yoshida, K. Koga, A. Hyodo, H. Ohtsuka, A. Kuniyasu, N. Tamamaki, Y. Sugimoto, and H. Nakayama. 2012. Intracerebral microinjection of interleukin-4/interleukin-13 reduces β-amyloid accumulation in the ipsilateral side and improves cognitive deficits in young amyloid precursor protein 23 mice. Neuroscience 207: 243–260.PubMedCrossRef Kawahara, K., M. Suenobu, A. Yoshida, K. Koga, A. Hyodo, H. Ohtsuka, A. Kuniyasu, N. Tamamaki, Y. Sugimoto, and H. Nakayama. 2012. Intracerebral microinjection of interleukin-4/interleukin-13 reduces β-amyloid accumulation in the ipsilateral side and improves cognitive deficits in young amyloid precursor protein 23 mice. Neuroscience 207: 243–260.PubMedCrossRef
33.
Zurück zum Zitat Liu, X., N. Jiang, and W. Zhou. 2023. Various energetic metabolism of microglia in response to different stimulations. Molecules 28. Liu, X., N. Jiang, and W. Zhou. 2023. Various energetic metabolism of microglia in response to different stimulations. Molecules 28.
34.
Zurück zum Zitat Xu, J., Z. Chen, F. Yu, H. Liu, C. Ma, D. Xie, X. Hu, R.K. Leak, S.H.Y. Chou, R.A. Stetler, et al. 2020. IL-4/STAT6 signaling facilitates innate hematoma resolution and neurological recovery after hemorrhagic stroke in mice. Proceedings of the National Academy of Sciences U S A 117: 32679–32690.CrossRef Xu, J., Z. Chen, F. Yu, H. Liu, C. Ma, D. Xie, X. Hu, R.K. Leak, S.H.Y. Chou, R.A. Stetler, et al. 2020. IL-4/STAT6 signaling facilitates innate hematoma resolution and neurological recovery after hemorrhagic stroke in mice. Proceedings of the National Academy of Sciences U S A 117: 32679–32690.CrossRef
35.
Zurück zum Zitat Jiang, Y., D.W. Liu, X.Y. Han, Y.N. Dong, J. Gao, B. Du, L. Meng, and J.G. Shi. 2012. Neuroprotective effects of anti-tumor necrosis factor-alpha antibody on apoptosis following subarachnoid hemorrhage in a rat model. Journal of Clinical Neuroscience 19: 866–872.PubMedCrossRef Jiang, Y., D.W. Liu, X.Y. Han, Y.N. Dong, J. Gao, B. Du, L. Meng, and J.G. Shi. 2012. Neuroprotective effects of anti-tumor necrosis factor-alpha antibody on apoptosis following subarachnoid hemorrhage in a rat model. Journal of Clinical Neuroscience 19: 866–872.PubMedCrossRef
36.
Zurück zum Zitat Ma, L., Y. Jiang, Y. Dong, J. Gao, B. Du, and D. Liu. 2018. Anti-TNF-alpha antibody attenuates subarachnoid hemorrhage-induced apoptosis in the hypothalamus by inhibiting the activation of Erk. Neuropsychiatric Disease and Treatment 14: 525–536.PubMedPubMedCentralCrossRef Ma, L., Y. Jiang, Y. Dong, J. Gao, B. Du, and D. Liu. 2018. Anti-TNF-alpha antibody attenuates subarachnoid hemorrhage-induced apoptosis in the hypothalamus by inhibiting the activation of Erk. Neuropsychiatric Disease and Treatment 14: 525–536.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Fragata, I., A. Bustamante, A. Penalba, P. Ferreira, A.P. Nunes, P. Canhão, and J. Montaner. 2020. TNF-R1 correlates with cerebral perfusion and acute ischemia following subarachnoid hemorrhage. Neurocritical Care 33: 679–687.PubMedCrossRef Fragata, I., A. Bustamante, A. Penalba, P. Ferreira, A.P. Nunes, P. Canhão, and J. Montaner. 2020. TNF-R1 correlates with cerebral perfusion and acute ischemia following subarachnoid hemorrhage. Neurocritical Care 33: 679–687.PubMedCrossRef
38.
Zurück zum Zitat Wang, X., D. Wen, C. You, and L. Ma. 2022. Identification of the key immune-related genes in aneurysmal subarachnoid hemorrhage. Frontiers in Molecular Neuroscience 15: 931753.PubMedPubMedCentralCrossRef Wang, X., D. Wen, C. You, and L. Ma. 2022. Identification of the key immune-related genes in aneurysmal subarachnoid hemorrhage. Frontiers in Molecular Neuroscience 15: 931753.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Herz, J., P. Sabellek, T.E. Lane, M. Gunzer, D.M. Hermann, and T.R. Doeppner. 2015. Role of neutrophils in exacerbation of brain injury after focal cerebral ischemia in hyperlipidemic mice. Stroke 46: 2916–2925.PubMedPubMedCentralCrossRef Herz, J., P. Sabellek, T.E. Lane, M. Gunzer, D.M. Hermann, and T.R. Doeppner. 2015. Role of neutrophils in exacerbation of brain injury after focal cerebral ischemia in hyperlipidemic mice. Stroke 46: 2916–2925.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Brait, V.H., J. Rivera, B.R. Broughton, S. Lee, G.R. Drummond, and C.G. Sobey. 2011. Chemokine-related gene expression in the brain following ischemic stroke: No role for CXCR2 in outcome. Brain Research 1372: 169–179.PubMedCrossRef Brait, V.H., J. Rivera, B.R. Broughton, S. Lee, G.R. Drummond, and C.G. Sobey. 2011. Chemokine-related gene expression in the brain following ischemic stroke: No role for CXCR2 in outcome. Brain Research 1372: 169–179.PubMedCrossRef
41.
Zurück zum Zitat Xie, W., T. Huang, Y. Guo, Y. Zhang, W. Chen, Y. Li, C. Chen, and P. Li. 2023. Neutrophil-derived cathelicidin promotes cerebral angiogenesis after ischemic stroke. Journal of Cerebral Blood Flow & Metabolism 271678x231175190. Xie, W., T. Huang, Y. Guo, Y. Zhang, W. Chen, Y. Li, C. Chen, and P. Li. 2023. Neutrophil-derived cathelicidin promotes cerebral angiogenesis after ischemic stroke. Journal of Cerebral Blood Flow & Metabolism 271678x231175190.
42.
Zurück zum Zitat Vallès, A., L. Grijpink-Ongering, F.M. de Bree, T. Tuinstra, and E. Ronken. 2006. Differential regulation of the CXCR2 chemokine network in rat brain trauma: Implications for neuroimmune interactions and neuronal survival. Neurobiology of Diseases 22: 312–322.CrossRef Vallès, A., L. Grijpink-Ongering, F.M. de Bree, T. Tuinstra, and E. Ronken. 2006. Differential regulation of the CXCR2 chemokine network in rat brain trauma: Implications for neuroimmune interactions and neuronal survival. Neurobiology of Diseases 22: 312–322.CrossRef
43.
Zurück zum Zitat Bonecchi, R., F. Facchetti, S. Dusi, W. Luini, D. Lissandrini, M. Simmelink, M. Locati, S. Bernasconi, P. Allavena, E. Brandt, et al. 2000. Induction of functional IL-8 receptors by IL-4 and IL-13 in human monocytes. The Journal of Immunology 164: 3862–3869.PubMedCrossRef Bonecchi, R., F. Facchetti, S. Dusi, W. Luini, D. Lissandrini, M. Simmelink, M. Locati, S. Bernasconi, P. Allavena, E. Brandt, et al. 2000. Induction of functional IL-8 receptors by IL-4 and IL-13 in human monocytes. The Journal of Immunology 164: 3862–3869.PubMedCrossRef
44.
Zurück zum Zitat Pattanaik, K.P., G. Ganguli, S.K. Naik, and A. Sonawane. 2021. Mycobacterium tuberculosis EsxL induces TNF-α secretion through activation of TLR2 dependent MAPK and NF-κB pathways. Molecular Immunology 130: 133–141.PubMedCrossRef Pattanaik, K.P., G. Ganguli, S.K. Naik, and A. Sonawane. 2021. Mycobacterium tuberculosis EsxL induces TNF-α secretion through activation of TLR2 dependent MAPK and NF-κB pathways. Molecular Immunology 130: 133–141.PubMedCrossRef
45.
Zurück zum Zitat Gea-Sorlí, S., and D. Closa. 2009. In vitro, but not in vivo, reversibility of peritoneal macrophages activation during experimental acute pancreatitis. BMC Immunology 10: 42.PubMedPubMedCentralCrossRef Gea-Sorlí, S., and D. Closa. 2009. In vitro, but not in vivo, reversibility of peritoneal macrophages activation during experimental acute pancreatitis. BMC Immunology 10: 42.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Prendiville, J., N. Thatcher, M. Lind, R. McIntosh, A. Ghosh, P. Stern, and D. Crowther. 1993. Recombinant human interleukin-4 (rhu IL-4) administered by the intravenous and subcutaneous routes in patients with advanced cancer–a phase I toxicity study and pharmacokinetic analysis. European Journal of Cancer 29a: 1700–1707.PubMedCrossRef Prendiville, J., N. Thatcher, M. Lind, R. McIntosh, A. Ghosh, P. Stern, and D. Crowther. 1993. Recombinant human interleukin-4 (rhu IL-4) administered by the intravenous and subcutaneous routes in patients with advanced cancer–a phase I toxicity study and pharmacokinetic analysis. European Journal of Cancer 29a: 1700–1707.PubMedCrossRef
47.
Zurück zum Zitat Enam, S.F., S.R. Kader, N. Bodkin, J.G. Lyon, M. Calhoun, C. Azrak, P.M. Tiwari, D. Vanover, H. Wang, P.J. Santangelo, and R.V. Bellamkonda. 2020. Evaluation of M2-like macrophage enrichment after diffuse traumatic brain injury through transient interleukin-4 expression from engineered mesenchymal stromal cells. Journal of Neuroinflammation 17: 197.PubMedPubMedCentralCrossRef Enam, S.F., S.R. Kader, N. Bodkin, J.G. Lyon, M. Calhoun, C. Azrak, P.M. Tiwari, D. Vanover, H. Wang, P.J. Santangelo, and R.V. Bellamkonda. 2020. Evaluation of M2-like macrophage enrichment after diffuse traumatic brain injury through transient interleukin-4 expression from engineered mesenchymal stromal cells. Journal of Neuroinflammation 17: 197.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Chen, X., J. Zhang, Y. Song, P. Yang, Y. Yang, Z. Huang, and K. Wang. 2020. Deficiency of anti-inflammatory cytokine IL-4 leads to neural hyperexcitability and aggravates cerebral ischemia-reperfusion injury. Acta Pharmaceutica Sinica B 10: 1634–1645.PubMedPubMedCentralCrossRef Chen, X., J. Zhang, Y. Song, P. Yang, Y. Yang, Z. Huang, and K. Wang. 2020. Deficiency of anti-inflammatory cytokine IL-4 leads to neural hyperexcitability and aggravates cerebral ischemia-reperfusion injury. Acta Pharmaceutica Sinica B 10: 1634–1645.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Brombacher, T.M., J.K. Nono, K.S. De Gouveia, N. Makena, M. Darby, J. Womersley, O. Tamgue, and F. Brombacher. 2017. IL-13-mediated regulation of learning and memory. The Journal of Immunology 198: 2681–2688.PubMedCrossRef Brombacher, T.M., J.K. Nono, K.S. De Gouveia, N. Makena, M. Darby, J. Womersley, O. Tamgue, and F. Brombacher. 2017. IL-13-mediated regulation of learning and memory. The Journal of Immunology 198: 2681–2688.PubMedCrossRef
Metadaten
Titel
Interleukin-4 Modulates Neuroinflammation by Inducing Phenotypic Transformation of Microglia Following Subarachnoid Hemorrhage
verfasst von
Jing Wang
Lili Wang
Qingjian Wu
Yichen Cai
Chengfu Cui
Ming Yang
Baoliang Sun
Leilei Mao
Yuan Wang
Publikationsdatum
29.10.2023
Verlag
Springer US
Erschienen in
Inflammation / Ausgabe 1/2024
Print ISSN: 0360-3997
Elektronische ISSN: 1573-2576
DOI
https://doi.org/10.1007/s10753-023-01917-z

Weitere Artikel der Ausgabe 1/2024

Inflammation 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Triglyzeridsenker schützt nicht nur Hochrisikopatienten

10.05.2024 Hypercholesterinämie Nachrichten

Patienten mit Arteriosklerose-bedingten kardiovaskulären Erkrankungen, die trotz Statineinnahme zu hohe Triglyzeridspiegel haben, profitieren von einer Behandlung mit Icosapent-Ethyl, und zwar unabhängig vom individuellen Risikoprofil.

Gibt es eine Wende bei den bioresorbierbaren Gefäßstützen?

In den USA ist erstmals eine bioresorbierbare Gefäßstütze – auch Scaffold genannt – zur Rekanalisation infrapoplitealer Arterien bei schwerer PAVK zugelassen worden. Das markiert einen Wendepunkt in der Geschichte dieser speziellen Gefäßstützen.

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Wie managen Sie die schmerzhafte diabetische Polyneuropathie?

10.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Mit Capsaicin-Pflastern steht eine neue innovative Therapie bei schmerzhafter diabetischer Polyneuropathie zur Verfügung. Bei therapierefraktären Schmerzen stellt die Hochfrequenz-Rückenmarkstimulation eine adäquate Option dar.

Update Innere Medizin

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