Skip to main content
Erschienen in: The Journal of Headache and Pain 1/2021

Open Access 01.12.2021 | Research article

Activation of microglial GLP-1R in the trigeminal nucleus caudalis suppresses central sensitization of chronic migraine after recurrent nitroglycerin stimulation

verfasst von: Feng Jing, Qian Zou, Yangyang Wang, Zhiyou Cai, Yong Tang

Erschienen in: The Journal of Headache and Pain | Ausgabe 1/2021

Abstract

Background

Central sensitization is considered a critical pathogenic mechanism of chronic migraine (CM). Activation of microglia in the trigeminal nucleus caudalis (TNC) contributes to this progression. Microglial glucagon-like peptide-1 receptor (GLP-1R) activation can alleviate pain; however, whether it is involved in the mechanism of CM has not been determined. Thus, this study aims to investigate the precise role of GLP-1R in the central sensitization of CM.

Methods

Repeated nitroglycerin injection-treated mice were used as a CM animal model in the experiment. To identify the distribution and cell localization of GLP-1R in the TNC, we performed immunofluorescence staining. Changes in the expression of GLP-1R, Iba-1, PI3K and p-Akt in the TNC were examined by western blotting. To confirm the effect of GLP-1R and PI3K/Akt in CM, a GLP-1R selective agonist (liraglutide) and antagonist (exendin(9–39)) and a PI3K selective antagonist (LY294002) were administered. Mechanical hypersensitivity was measured through von Frey filaments. To investigate the role of GLP-1R in central sensitization, calcitonin gene-related peptide (CGRP) and c-fos were determined using western blotting and immunofluorescence. To determine the changes in microglial activation, IL-1β and TNF-α were examined by western blotting, and the number and morphology of microglia were measured by immunofluorescence. We also confirmed the effect of GLP-1R on microglial activation in lipopolysaccharide-treated BV-2 microglia.

Results

The protein expression of GLP-1R was increased in the TNC after nitroglycerin injection. GLP-1R was colocalized with microglia and astrocytes in the TNC and was fully expressed in BV-2 microglia. The GLP-1R agonist liraglutide alleviated basal allodynia and suppressed the upregulation of CGRP, c-fos and PI3K/p-Akt in the TNC. Similarly, the PI3K inhibitor LY294002 prevented nitroglycerin-induced hyperalgesia. In addition, activating GLP-1R reduced Iba-1, IL-1β and TNF-α release and inhibited TNC microglial number and morphological changes (process retraction) following nitroglycerin administration. In vitro, the protein levels of IL-1β and TNF-α in lipopolysaccharide-stimulated BV-2 microglia were also decreased by liraglutide.

Conclusions

These findings suggest that microglial GLP-1R activation in the TNC may suppress the central sensitization of CM by regulating TNC microglial activation via the PI3K/Akt pathway.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s10194-021-01302-x.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BBB
Blood-brain barrier
CGRP
Calcitonin gene-related peptide
CM
Chronic migraine
CNS
Central nervous system
CSD
Cortical spreading depression
DMEM
Dulbecco’s modified Eagle’s medium
FBS
Foetal bovine serum
GLP-1R
Glucagon-like peptide-1 receptor
IGF-1
Insulin-like growth factor-1
IS
Inflammatory soup
LPS
Lipopolysaccharide
NTG
Nitroglycerin
PI3K
Phosphoinositide 3-kinase
PKA
Protein kinase A
PMSF
Phenylmethylsulfonyl fluoride
RT
Room temperature
TNC
Trigeminal nucleus caudalis

Background

Chronic migraine (CM) is a severe neurological disease that seriously affects the daily life of patients and imposes a large economic burden on society [1, 2]. CM is the most common chronic headache in headache clinics in China [3]. The chronic condition of CM is attributable to repeated migraine attacks, and 2.5 % of episodic migraine converts into CM each year [4, 5]. Therefore, early prophylactic treatment is necessary for improving the prognosis of CM patients. However, the present treatments for CM are not satisfactory [6], and the pathogenesis of CM is still not fully understood.
Accumulating evidence indicates that central sensitization is a critical mechanism of CM, which manifests as neuronal plastic changes in the trigeminal nucleus caudalis (TNC) [79]. Studies have suggested that microglia may participate in crosstalk with neurons through chemotaxis, phagocytosis, proinflammatory cytokine and neurotrophin production [1012]. In a cortical spreading depression (CSD) model [13, 14], which mimics the aura of migraine, and the chronic inflammatory soup (IS)-triggered CM model, microglia are dramatically activated [15, 16]. In addition, our previous studies revealed that microglia and their purinergic receptors (P2 × 4R, P2 × 7R and P2Y12R) were significantly upregulated in the TNC after NTG injection, and inhibiting microglial activation, including morphological and inflammatory changes, might affect neuronal hyperexcitability in the TNC, which ultimately relieved CM-associated allodynia [1719]. These findings demonstrate that TNC microglia play a crucial role in the central sensitization of CM, and a better understanding of the function of TNC microglia will help us to further understand the pathogenesis of CM.
Recently, Wang Yongxiang and colleagues provided the first evidence that activation of microglial glucagon-like peptide-1 receptor (GLP-1R) in the spinal cord specifically suppresses neuropathic pain, cancer pain, and diabetic hypersensitivity [20]. However, whether GLP-1R is involved in the central sensitization of CM has not yet been studied. GLP-1 is an endogenous insulinotropic hormone secreted from L cells of the small intestine, that participates in the homeostatic regulation of insulin and glucose by activating GLP-1R [21, 22]. GLP-1R is a kind of G-protein-coupled receptor. Activation of GLP-1R can inhibit apoptosis and inflammation by stimulating phosphoinositide 3-kinase (PI3K) and protein kinase A (PKA) [23]. GLP-1R expressed in the central nervous system (CNS) [24] may also be involved in regulating cell proliferation, neuronal excitability and synaptic plasticity [2527]. However, the distribution and function of GLP-1R in the TNC have not yet been examined. Previous studies have revealed that migraine patients suffer from impaired insulin sensitivity and have higher blood glucose levels [28]. In addition, insulin-like growth factor-1 (IGF-1) treatment can inhibit CSD and calcitonin-gene-related peptide (CGRP) production, thereby relieving migraine-related hyperalgesia [29]. These data combined with the effect of GLP-1R in chronic pain suggest that activation of GLP-1R may also alleviate CM.
Based on these results, we hypothesized that GLP-1R in the TNC mediates microglial activation via the PI3K/Akt pathway and suppresses the central sensitization of CM. In the present study, we investigated whether activation of GLP-1R prevented allodynia and inhibited microglial morphological changes and inflammation in the TNC using a chronic nitroglycerin (NTG) injection-stimulated mouse model and the selective GLP-1R agonist liraglutide. We also confirmed the role of GLP-1R on microglia in lipopolysaccharide (LPS)-incubated BV-2 cells. In addition, we examined the involvement of the PI3K/Akt pathway in CM. Our findings suggest for the first time that TNC microglial GLP-1R activation suppresses the central sensitization of CM and that GLP-1R may serve as a new target for treating CM.

Materials and methods

Animals

Male C57BL/6 mice weighing 18–20 g were kindly provided by the Experimental Animal Center of Chongqing Medical University (Chongqing, China). Animal experiments were conducted in accordance with Chongqing Medical University Animal Ethics Committee and were approved by the National Institutes of Health guidelines on animal care. Mice were housed with a 12 h light/dark cycle, at a stable temperature of 23 ± 2 °C and humidity of 50 ± 10 %, and were given food and water ad libitum. Every effort was made to minimize the number of mice used in the experiments and their suffering.

Drug treatments

Nitroglycerin (NTG) (5.0 mg/ml, Henan Reagent, China) was freshly diluted to a final concentration of 1 mg/ml with saline, containing 6 % propylene glycol and 6 % alcohol. The intraperitoneal (i.p.) delivery of the diluted NTG was performed as described previously, that is, injected at a dose of 10 mg/kg every other day for 9 d (i.e., days 1, 3, 5, 7, and 9). Mice in the sham group were injected with an equivalent volume of saline, 6 % propylene glycol, and 6 % alcohol.
To measure the effect of GLP-1R in CM, a selective GLP-1R agonist (liraglutide) and an antagonist (exendin(9–39)) were applied in the experiment. Liraglutide (800 µg/kg; Selleck, TX, USA) and exendin(9–39) (50 µg/kg; MedChemExpress/MCE, USA), diluted with sterile saline, were administered i.p. for 16 consecutive days; that is, these drugs were administered 1 week before NTG injection. The time point for injections was also selected before NTG administration. To examine the role of PI3K/Akt in CM, the PI3K/Ak antagonist LY294002 (20 mg/kg; Selleck, TX, USA), diluted with sterile saline, was injected i.p. five times every other day prior to NTG injections. The dose and delivery method of these drugs were based on previous research [3033] and our experience.

Body weight and Blood glucose test

After a week of adaptation, the animals were randomly assigned to different experimental groups, as shown in Table 1 in which the experimental sets and sample size were delineated. During the whole experiment, the body weight and blood glucose of mice were measured 3 times, respectively, before drug administration, 1 week and 16 d after delivering liraglutide or exendin(9–39). The time point was selected at 9:00 prior to NTG injection. Glucose measurement was performed by a blood glucose meter (Sinocare, Changsha, China) using blood collected from the tail vein of mice.
Table 1
Schematic representation of experimental groups, sample size (number, n) and the sampling time (hour, h) of mice per group for different experimental analysis
Experimental groups
Total (n)
Behavioural test (n)
WB (other) (n)
WB (c-fos) (n)
IF (other) (n)
IF (c-fos) (n)
Sampling time (h)
Sham
9
-
6
-
3
-
24h after the first, the second, the third and the last NTG i.p. injections
NTG (1d)
6
-
6
-
-
-
NTG (3d)
6
-
6
-
-
-
NTG (5d)
6
-
6
-
-
-
NTG (9d)
9
-
6
-
3
-
Sham
20
8
6
6
4
4
For c-fos: 2h after the last NTG/saline i.p. injection;
For other targets: 24h after the last NTG/saline i.p. injection
Exe(9-39)
12
8
6
6
-
-
Lira
12
8
6
6
-
-
CM
20
8
6
6
4
4
CM+Exe(9-39)
20
8
6
6
4
4
CM+Lira
20
8
6
6
4
4
CM+LY294002
8
8
-
-
-
-
WB (other), western blot for GLP-1, GLP-1R, CGRP, Iba-1, IL-1β, TNF-α, PI3K and p-AKT; IF (other): immunofluorescence for GLP-1R, CGRP, GFAP, NeuN and Iba-1; Exe(9-39), exendin(9-39); Lira, liraglutide

BV-2 cell culture and treatment

BV-2 cells were provided by Procell Life Science & Technology (Wuhan, China). Cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco, NY, USA) supplemented with 10 % foetal bovine serum (FBS) (Gibco, NY, USA) and were maintained in a humidified incubator at 37 °C and 5 % CO2.
Cells were incubated with serum-free media for 24 h prior to each experiment and seeded at 1 × 106 cells/dish in 100 mm culture dishes for western blotting and 3 × 104 cells/well in 24-well- culture plates for immunofluorescence. To study the role of GLP-1R in microglial inflammation, BV-2 cells were preincubated with the selective GLP-1R agonist liraglutide (100 nM, Selleck, TX, USA) and the antagonist exendin(9–39) (200 nM, MCE, USA) for 24 h, followed by LPS (1 µg/ml, Sigma–Aldrich, MO, USA) stimulation for 24 h. The doses of these drugs used with cells were based on previous data [3436].

Behavioural tests

Mice were acclimated to the testing chambers for 3 successive days before the behavioural tests. All experiments were performed in a low-light room between 9:00 and 15:00 at time points before and 2 h after NTG injection. The experimenter was blinded to the dug condition and the behavioural data analysis.
Both head-specific (periorbital) and hindpaw hyperalgesia were measured to reveal the hypersensitivity of the CM animal model. Mechanical responses were detected by a set of von Frey filaments (bending force ranging from 0.008 to 2 g, Aesthesio, USA) according to the up and down method [37, 38]. The first filament was chosed as 0.4 g. For periorbital testing, mice were placed in a paper cup to which they had previously been habituated, and then the periorbital region, positioned caudal to the eyes and near the midline, was stimulated with a von Frey filament. A positive response was considered quick retraction of the head or scratching of the face with the ipsilateral forepaw. In the absence of a response, a heavier filament (up) was applied, and in the presence of a response, a lighter filament (down) was applied. The duration of each stimulus was 3 s with an interval of at least 1 min. This process was followed for a maximum of 4 filaments after the first response. For hindpaw sensitivity, mice were habituated to acrylic cages 30 min before the test, and then the central area of the plantar surface of the hindpaw was stimulated with von Frey filaments. Positive responses were defined as lifting, shaking or licking of the paw. Testing was performed according to the up-down method described above.

Western blotting

To analyze the protein expression of GLP-1, GLP-1R, PI3K and p-Akt on different days after NTG intermittent injection, TNC samples were collected 24 h after the first (NTG 1d), the second (NTG 3d), the third (NTG 5d) and the last (NTG 9d) NTG injections. To examine c-fos expression, TNC tissues were collected 2 h after the last NTG injection, while for other protein targets, TNC was isolated and collected 24 h after the last NTG administration. The sampling time was shown in Table 1. Tissues were homogenized in cold RIPA lysis buffer (Beyotime, Shanghai, China) containing phenylmethylsulfonyl fluoride (PMSF, Beyotime, Shanghai, China) at 4 °C for 1 h. The total amount of protein was determined using a BCA protein assay kit (Beyotime, Shanghai, China). The protein samples (40 µg) were separated by 10 % SDS-polyacrylamide gel (Beyotime, Shanghai, China) electrophoresis and transferred to PVDF membranes. Following transfer, the membranes were blocked in TBST containing 5 % skim milk for 2 h at room temperature (RT). Then, the blots were incubated with the following antibodies overnight at 4 °C: mouse anti-GLP-1R (1:200, Santa Cruz, CA, USA), mouse anti-GLP-1 (1:100, Santa Cruz, CA, USA), mouse anti-CGRP (1:100, Santa Cruz, CA, USA), mouse anti-c-fos (1:200, Santa Cruz, CA, USA), rabbit anti-Iba-1 (1:1000, Abcam, Cambridge, MA, USA), rabbit anti-PI3K (1:1000, Proteintech, China), rabbit anti-Akt (1:1000, Proteintech, China), rabbit anti-phospho-Akt (1:2000, Cell Signalling, Boston, MA, USA), rabbit anti-IL-1β (1:800, Wanleibio, China), rabbit anti-TNF-α (1:800, Wanleibio, China), and mouse anti-β-actin (1:1000, Beyotime, China). After the membranes had been washed with TBST, the secondary antibodies (goat anti-rabbit, 1:1000; goat anti-mouse, 1:1000; Beyotime, China) were applied and incubated for 1 h at RT. After the secondary antibody reaction, the bands were visualized with enhanced chemiluminescence, and the positive pixel area was detected by an image analysis system (Fusion, Germany). The band sensitivities were normalized against the corresponding β-actin loading controls. The western blotting test was repeated six times for each target, and consistent results were obtained.
To investigate the expression level of various proteins in BV-2 cells, we collected cells after LPS incubation. Cells were lysed in cold RIPA buffer containing PMSF for 30 min at 4 °C, and the total protein concentration was determined using a BCA protein assay kit. The following procedures were performed according to the method described above. Each analysis was repeated in three independent experiments.

Immunofluorescence staining and counting

To stained c-fos, TNC tissues were collected 2 h after the last NTG injection, while for staining other targets, TNC was collected 24 h after the last NTG administration. Mice were deeply anaesthetized and perfused transcardially with 30 ml of PBS (pH 7.4) followed by 30 ml of cold 4 % paraformaldehyde (PFA) in PBS. The whole brain and cervical spinal cord (C1-C2) were collected and postfixed with 4 % PFA overnight at 4 °C.The medullary segment including the TNC between + 1 and − 3 mm from the obex was removed and transferred to 30 % sucrose for 48 h. After being frozen at − 80 °C, samples were cut into 10-µm-thick sections on a cryostat (Thermo). Sample sections were blocked in 5 % goat serum with 0.3 % Triton X-100 for 1 h at RT and then incubated overnight at 4 °C with the following primary antibodies: mouse anti-GLP-1R (1:50, Santa Cruz, CA, USA), mouse anti-GLP-1(1:50, Santa Cruz, CA, USA), rabbit anti-Iba1 (1:500, Wako Chemicals, Tokyo, Japan), rabbit anti-GFAP (1:200, Cell Signalling, Boston, MA, USA), rabbit anti-NeuN (1:100, Proteintech, China), mouse anti-CGRP (1:100, Santa Cruz, CA, USA), and mouse anti-c-fos (1:100, Santa Cruz, CA, USA). The sections were then incubated with fluorescence-conjugated secondary antibodies (Alexa Fluor Cy3 and Alexa Fluor 488, 1:500, Beyotime, China). Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI) (Beyotime, China) at RT for 10 min. The sections were visualized and photographed with a confocal microscope (TCS Sp8, Leica). Quantification of the fluorescent signal intensity was performed by image analysis software (Image-Pro Plus 6.2, Media Cybernetics).
To quantify the immunoreactivity of CGRP and the numbers of c-fos- and iba-1- positive cells in the TNC, 6 sections per mouse from 4 mice were selected randomly for each group, and 6–8 visual fields per section were captured. Images centred on the superficial layer of the TNC were captured under a ×100 or × 200 objective, and the immunoreactive cells in this region were counted with Image-Pro Plus.
To quantify the length of microglial processes, 6–8 visual fields per section were selected from 6 sections per mouse (n = 4), and all intact cells in the field of view were measured. Skeletal images of each cell were drawn by Neuron J (an ImageJ plug-in), and then the total and mean lengths of microglial processes were automatically determined by the software.

Statistical analysis

Statistical analyses were performed by SPSS 22.0 software (IBM Corp, Armonk, NY, USA). All data are expressed as mean ± SEM. The statistical significance of changes between the values was determined by one-way ANOVA with a post hoc test (Tukey’s test). Behavioural data were evaluated by two-way ANOVA with a Bonferroni post hoc test. P < 0.05 was considered to be statistically significant.

Results

Upregulation of GLP-1R in the TNC after chronic NTG injection

To examine whether GLP-1R was expressed in the TNC, we performed immunofluorescence for GLP-1R using a specific antibody, and our images showed that GLP-1R was specifically expressed in the TNC in normal mice (Fig. 1a). To study the effect of NTG on GLP-1R activity, we used western blotting to detect the level of GLP-1R in the TNC and found that the expression of GLP-1R was increased after chronic NTG injection in a time-dependent manner (Fig. 1b) and peaked on day 9 (0.74 ± 0.13) (Fig. 1c, p < 0.05). In addition, we detected the protein level of GLP-1 in the TNC after NTG administration, which is a natural agonist of GLP-1R. We observed that recurrent NTG injection induced a time-dependent decrease in GLP-1 levels in the immunoblot analyses (Fig. 1b). GLP-1 expression was significantly decreased on day 5 (0.56 ± 0.18) and day 9 (0.60 ± 0.14) (Fig. 1d, p < 0.01) after NTG injection. We also performed immunofluorescence and observed that the number of GLP-1R-positive cells in the NTG (9d) group was higher than that in the sham group (Fig. 1e). These results indicate that GLP-1R is dramatically upregulated in the TNC in NTG-induced CM model.

Localization of GLP-1R in the TNC

To localize the specific cell type for GLP-1R in the TNC, immunofluorescence was stained with Ibal-1 (microglial marker), GFAP (astrocytic marker) and NeuN (neuronal marker) using TNC sections in the CM group (Fig. 2a). Our images showed that GLP-1R-positive cells were mainly double labelled with Iba-1 and GFAP but did not colocalize with NeuN (Fig. 2b). These results suggest that GLP-1Rs exclusively localize to glial cells but not to neuronal cells.

Activation of GLP-1R attenuated CM-associated mechanical allodynia

Considering the significant upregulation of GLP-1R in the TNC and the widely recognized effect of microglia in CM, it is reasonable to expect that GLP-1R may play an important role in the pathogenesis of CM. To study whether GLP-1R activation in TNC microglia can alleviate NTG-induced allodynia, the selective GLP-1R agonist liraglutide (800 µg/kg, n = 8 per group) and the antagonist exendin(9–39) (50 µg/kg, n = 8 per group) were i.p. injected for 16 d (Fig. 3a). The injection of the drugs started 1 week before NTG administration.
Liraglutide is a commonly used hypoglycaemic agent and is known to improve glucose homeostasis in diabetic patients [39]. To avoid the influence of hypoglycaemia on the experiment, we evaluated the changes in blood glucose and body weight of the experimental mice (Fig. 3b-c). The results showed that there was no significant difference in the levels of blood glucose in the various groups (Fig. 3b). Interestingly, the body weight of the CM combined with liraglutide group was significantly reduced compared with that of the sham group (Fig. 3c).
The periorbital and paw withdrawal thresholds were measured every other day from 1 to 9 d after NTG injection. The basal responses were evaluated before NTG, and the post-treatment responses were evaluated 2 h after NTG administration (Fig. 3d-g). We found that liraglutide (CM + Lira) markedly alleviated basal periorbital and paw hypersensitivity (p < 0.05). Chronic exendin(9–39) (CM + Exe(9–39)) injection did not improve CM-associated allodynia. Only liraglutide and exendin(9–39) injection did not provoke a significant effect on mechanical hypersensitivity.

Activation of GLP-1R suppressed NTG-induced CGRP expression in the TNC

CGRP is a widely recognized migraine-related mediator and is also involved in central sensitization in the TNC. To further confirm the effect of GLP-1R on central sensitization of CM, we measured alterations in CGRP expression in different drug-treated groups. The western blotting results showed that the protein level of CGRP in the TNC was significantly increased in the CM (p < 0.001) and GLP-1R antagonist exendin(9–39) treated groups (p < 0.05, Exe(9–39) vs. sham; p < 0.001, CM + Exe(9–39) vs. sham) (Fig. 4a-b). Activation of GLP-1R by its agonist liraglutide (CM + Lira) markedly suppressed the expression of CGRP compared with the corresponding level in the CM group (Fig. 4b, 0.58 ± 0.26 vs. 1.08 ± 0.12, p < 0.01). In addition, we performed immunofluorescence assessment of CGRP in the TNC (Fig. 4d). The density of CGRP-immunoreactive fibres in the TNC was increased in the CM and exendin(9–39) treated (CM + Exe(9–39)) groups compared with the sham group (Fig. 4c, p < 0.001). Consistent with the changes in CGRP levels in immunoblot analyses, the immunoreactivity of CGRP in the TNC in the CM group was significantly decreased by liraglutide treatment (Fig. 4c, p < 0.001).

Activation of GLP-1R reduced NTG-induced c-fos expression in the TNC

In addition to CGRP, we also detected alterations in c-fos expression in the TNC, which is a reliable marker of neuronal activation and is considered another mediator of central sensitization. Using western blotting, we found that the protein levels of c-fos in the TNC in the CM (1.13 ± 0.14) and exendin(9–39) treated group (1.08 ± 0.20) were significantly upregulated (Fig. 5a-b, p < 0.001 CM vs. Sham; p < 0.01 CM + Exe(9–39) vs. Sham), and liraglutide treatment markedly suppressed the expression of c-fos in CM mice (Fig. 5a-b, 0.72 ± 0.14, p < 0.01 CM + Lira vs. Sham). Consistent with the changes of c-fos levels in immunoblot analyses, the immunofluorescence results for c-fos in the TNC showed that the number of c-fos immunoreactive cells in the exendin(9–39) treated group (CM + Exe(9–39), 72.33 ± 14.95) was higher than that of sham group (39.83 ± 13.23) (Fig. 5c-d, p < 0.01); however, the number of c-fos- positive cells in the liraglutide treated group (CM + Lira, 51 ± 11.22) was significantly decreased compared with that in the CM group (72.67 ± 10.69) (Fig. 5c-d, p < 0.05).

GLP-1R contributes to microglial activation in the TNC in NTG-induced CM mice

We have indicated that chronic NTG stimulation induces an increase in the number and activity of microglia in the TNC. To study the involvement of GLP-1R in these changes, we measured microglia from TNC slices of different drug-treated groups using immunofluorescence. Quantification of Iba1-positive cells revealed that the number of microglia in the CM (272.6 ± 47.44) and exendin(9–39) treated group (CM + Exe (9–39), 269.2 ± 19.04) was significantly increased compared with that in the sham group (180.8 ± 26.41) (Fig. 6a,e, p < 0.01). The agonist liraglutide markedly suppressed the NTG-induced upregulation of microglia in the TNC (Fig. 6a, e, CM + Lira 212.6 ± 26.92, p < 0.05). We observed that the activated microglia in the TNC in CM mice manifested as hypertrophic somata and shortened processes; thus, we quantified the microglial processes in the highly magnified stacked images of Iba1-immunoreactive cells in the TNC (Fig. 6b-c). In addition, skeletal images of microglia in various groups were drawn for accurate quantification (Fig. 6d). The results showed that GLP-1R agonist liraglutide treatment partially reversed the NTG-induced shortening of both total (100.45 ± 40.55 μm vs. 67.99 ± 31.03 μm, p < 0.01) and mean lengths (20.28 ± 8.11 μm vs. 14.63 ± 5.20 μm, p < 0.01) of microglial processes in the TNC (Fig. 6f-g).
In addition, we examined the protein level of Iba-1 in the TNC using western blotting and found that liraglutide markedly reduced NTG-induced upregulation of Iba-1 (CM + Lira 0.76 ± 0.12 vs. CM 1.01 ± 0.14, p < 0.05) (Fig. 6h-i). We also detected the expression of the microglial proinflammatory factors IL-1β and TNF-α in the TNC during chronic NTG administration. The results showed that the protein levels of both IL-1β (p < 0.01) and TNF-α (p < 0.001) were increased after recurrent NTG and exendin(9–39) injection (Fig. 6h, j-k). However, the release of IL-1β and TNF-α in the TNC in CM mice was significantly reversed by chronic liraglutide treatment (Fig. 6h, j-k, p < 0.01). Together, these results indicate that GLP-1R activity contributes to alterations in the number, morphology and inflammatory factor production of microglia in the TNC following NTG administration.

Effect of GLP-1R on LPS-stimulated BV-2 microglial inflammatory cytokine release

Because liraglutide and exendin(9–39) were administered systematically in vivo, to further determine the effect of GLP-1R on microglial function, we carried out experiments in BV-2 microglia. Using double-labelling immunofluorescence, we observed that GLP-1R was colocalized with Iba1 in BV-2 cells (Fig. 7a). After LPS stimulation, the expression of GLP-1R was significantly increased compared with that of the sham group (Fig. 7b-c, 0.89 ± 0.06 vs. 0.59 ± 0.06, p < 0.01). Exendin(9–39) incubation partially reversed the expression of GLP-1R induced by LPS. However, the agonist liraglutide induced higher upregulation of GLP-1R compared with that in the LPS group. (Fig. 7b-c, 1.08 ± 0.19 vs. 0.89 ± 0.06, p < 0.05). We also quantified the expression of Iba-1, IL-1β and TNF-α after drug treatment, and the results showed that liraglutide reduced the protein level of Iba-1 (0.40 ± 0.11 vs. 0.68 ± 0.13, p < 0.01) and reversed the production of IL-1β (0.38 ± 0.09 vs. 0.63 ± 0.13, p < 0.01) and TNF-α (0.52 ± 0.09 vs. 0.72 ± 0.10, p < 0.05) induced by LPS (Fig. 7b, d-f). These results suggest that microglial GLP-1R mediates the inflammatory factor release of BV-2 microglia.

GLP-1R is required for PI3K/Akt activation in the TNC following recurrent NTG injection

Using western blotting, we measured the protein expression of PI3K and p-Akt in the TNC after repeated NTG injection (1, 3, 5, and 9 d post-injection) and found that these protein levels gradually increased in a time-dependent manner and peaked on day 9 (p < 0.05, PI3K in NTG 9d, 1.05 ± 0.18 vs. Sham 0.66 ± 0.17; p < 0.05, p-Akt in NTG 9d 0.69 ± 0.19 vs. Sham 0.40 ± 0.09) (Fig. 8a-c). Following recurrent treatment with the GLP-1R agonist liraglutide (CM + Lira), we observed a significant decrease in the expression of PI3K (0.68 ± 0.05 vs. 1 ± 0.18, p < 0.01) and p-Akt (0.42 ± 0.07 vs. 0.79 ± 0.12, p < 0.001) (Fig. 8d-f). The GLP-1R antagonist exendin(9–39) had no effect on NTG-induced PI3K and p-Akt expression levels. These results reveal that GLP-1R regulates PI3K/p-Akt activity in the TNC.

Inhibition of PI3K/Akt activity alleviated NTG-induced mechanical hypersensitivity

To evaluate whether PI3K/ Akt is involved in NTG-induced pain hypersensitivity, we i.p. injected the PI3K antagonist LY294002 (20 mg/kg) 5 times prior to NTG injection every other day (n = 8 per group). The behavioural results showed that repeated LY294002 treatment partially reversed basal periorbital and paw hyperalgesia (Fig. 8g-j), which was markedly attenuated on days 7 and 9. However, LY294002 administration had no significant improvement in post-treatment allodynia induced by NTG. These data indicate that PI3K/Akt, downstream of GLP-1R, contributes to the development of CM.

Discussion

In the current study, we observed the following new findings. (1) Chronic NTG injection induced a dramatic increase in GLP-1R in TNC microglia. (2) Activation of GLP-1R attenuated CM-associated allodynia and reduced the expression of CGRP and c-fos. (3) Activation of GLP-1R suppressed microglial morphological changes and inflammatory factor release in the TNC in CM mice and in vitro. (4) The PI3K/Akt pathway, inhibited by GLP-1R in the TNC, was proven to participate in the development of CM.
Previous studies have found that GLP-1Rs are expressed in various tissues, including the pancreatic islets, brain, dorsal root ganglia, and spinal cord [20, 4043]. Regarding the cellular localization of GLP-1R in the CNS, there are controversial findings. In the cerebral cortex, GLP-1R is expressed on neurons and glia [42], while in the spinal cord, GLP-1R is coexpressed only with microglia [20]. To date, there have been no studies on the distribution and localization of GLP-1R in the TNC. For the first time, we investigated that GLP-1R was widely expressed in the TNC and was specifically localized to microglia and astrocytes using immunofluorescence staining. Our western blotting data showed that GLP-1R protein expression in the TNC was increased after recurrent NTG injection. However, there is a major limitation that our present study includes only male animals. We have indicated that male mice could also successfully establish NTG induced CM model [17, 18], it is well known that sex differences in migraine is mainly due to the role of estrogen [44, 45], thus, to avoid the influence of estrogen, we selected male mice for further study. Whether the distribution and activation of TNC GLP-1R in female animals are different remains to be further studied.
We have indicated that the number of microglia in the TNC is also increased in this study and our previous studies [17, 18]. However, we did not find a significant upregulation of astrocytes in the TNC in this NTG-induced CM model in our previous researches. Considering the consistency of the alterations in GLP-1R and microglia in the TNC, we predict that the main reason for the upregulation of GLP-1R is due to the increased number of microglia. Consistent with our data, the upregulation of GLP-1R in the spinal cord on activated microglia was also found after peripheral nerve injury [20]. However, data on the changes in GLP-1R levels in other CNS disorders accompanied by microglial activation are contradictory. For example, the expression of GLP-1R in Alzheimer’s disease, experimental autoimmune encephalomyelitis and depression [4648] was found to be decreased in the brain. The differences in GLP-1R expression in activated microglia might be due to the different regions of the CNS and the various methods for activating microglia. Together, our data reveal that activating TNC microglia may increase the expression of GLP-1R in CM mice in response to chronic NTG injection.
Our behavioural results showed that chronically activating GLP-1R by means of consecutive i.p. injections of the selective agonist liraglutide attenuated NTG-induced basal tactile allodynia. However, blocking GLP-1R did not improve CM-related hypersensitivity. This finding suggests that liraglutide mainly produces prophylactic analgesic effects. Liraglutide, a GLP-1 analogue, is a new therapeutic drug for the treatment of diabetes. The drug can cross the blood-brain barrier (BBB) and activate GLP-1R in the CNS [49, 50], thus producing neuroprotective and anti-inflammatory effects [5055]. Previous data demonstrated that activation of GLP-1R could alleviate neuropathic pain, cancer pain and diabetic neuropathy [20]. In the present study, we provide the first evidence that liraglutide prevented the development of CM by activating GLP-1R in the TNC. It is known that CM shares a common mechanism of central sensitization with chronic pain; thus, our data complement the impact of GLP-1R on chronic pain.
It is well known that GLP-1Rs themselves are inactive and needs to be activated or inhibited to exert their function. Therefore, to clarify whether the function of GLP-1R is activated or inhibited in CM, we detected the changes in the level of endogenous GLP-1, a natural agonist of GLP-1R, in the TNC in NTG-induced CM mice. We performed western blotting to examine the protein level of endogenous GLP-1 in the TNC during repeated NTG administration and also conducted double-staining of GLP-1 and iba-1 to further detect the distribution of GLP-1 in the TNC. The results showed that GLP-1 was co-localized with microglia in the TNC, recurrent NTG administration decreased the number of GLP-1-positive cells compared with that in the sham group (Fig. S1). These results suggested that GLP-1 expression was gradually decreased in CM, and also revealed that although the protein expression of GLP-1R was upregulated after NTG stimulation, the amount of GLP-1Rs that can be activated was decreased, so the activity of GLP-1R in the TNC in CM model was actually inhibited. This data supports our behavioural analysis that activating GLP-1R by its agonist liraglutide plays a major role in relieving CM-related hyperalgesia.
Tactile allodynia is a clinical characteristics of CM patients which manifests as abnormal skin pain of the head and limbs [56], as we measured above (periorbital and paw responses); this is the external manifestation of central sensitization in the pathogenesis of CM. To further confirm the effect of GLP-1R on the central sensitization of CM, we examined the expression of CGRP and c-fos in the TNC. CGRP is a specific neuropeptide in the trigeminal system that plays a critical role in peripheral and central sensitization [9, 57]. Studies have indicated that NTG administration can induce the upregulation of CGRP in the TNC, dura mater, and blood [57]. C-fos, an immediate early gene that is widely considered a marker of neuronal activity, is also involved in central sensitization [58]. Studies in different animal models of migraine (NTG, CSD and IS stimulation) demonstrate a significant upregulation of c-fos in laminae I and II of the TNC [59]. Our findings showed that activating GLP-1R by i.p. injection of liraglutide dramatically suppressed the upregulation of CGRP and c-fos in the superficial lamina of the TNC induced by NTG. This result, combined with our behavioural data, confirms that activation of GLP-1R in the TNC may inhibit the central sensitization of CM.
Accumulating evidence has revealed that changes in microglial functions (for example, morphological changes, process movement, and inflammation) are required for the development of chronic pain [6062]. Our previous studies have indicated that microglial activation in the TNC contributes to the central sensitization of CM [17, 18]. Thus, we further explored whether GLP-1R is involved in CM by mediating microglial activation, including morphological changes and inflammation. In this study, chronic NTG injection induced an increase in microglial cell numbers and a significant reduction in microglial process length and inflammatory factor (IL-1β and TNF-α) release. Following liraglutide treatment, the upregulation of microglial cell number and proinflammatory factors was markedly decreased, and microglia exhibited much longer process lengths in the TNC. However, since the drugs were administered systemically in a simulated clinical manner, we cannot rule out the effect of GLP-1R in the peripheral system. Thus, we carried out experiments in BV-2 microglia to more accurately determine the effect of GLP-1R on microglial function. Consistent with the results in the CM animal model, GLP-1R is completely colocalized with BV-2 microglia, and activation of GLP-1R by liraglutide reduced Iba-1 protein expression and inflammatory factors. From these data, we conclude that GLP-1R is involved in regulating TNC microglial activation during NTG injection, and this function of GLP-1R might also be the mechanism for attenuating CM-related allodynia. Although our data in vivo and in vitro demonstrated that activating GLP-1R inhibited microglial activation, changes in microglia induced by liraglutide can be indirect as well as behavioral effects could be driven by other cells cause our treatments are delivered systemically. Because of the limit of this study, the potential role of GLP-1R in the TNC still needs to be further explored.
Studies have indicated that GLP-1R is involved in regulating cell proliferation and migration, neuronal activity, and inflammation through its downstream PI3K/Akt pathway [23, 42, 63]. Moreover, PI3K/Akt has been reported to be upregulated in migraine [64], but no study has provided a direct evidence of whether this pathway participates in the mechanism of CM. Our study confirmed that the PI3K/Akt pathway acts downstream of GLP-1R in the TNC in CM mice. In addition, we observed that the protein expression of PI3K and p-Akt in the TNC gradually increased in the CM model, and inhibiting the PI3K/Akt pathway by repeated administration of the selective antagonist LY294002 relieved NTG-induced basal tactile allodynia. These results provide the first evidence that inhibiting the PI3K/Akt pathway attenuates CM-associated allodynia; that is, the pathway participates in the central sensitization of CM. The involvement of GLP-1R in the pathogenesis of CM may also be realized by regulating the activity of this pathway.

Conclusions

Our study demonstrated that the expression of GLP-1R is increased in TNC microglia after NTG injection. Activation of GLP-1R with its selective agonist liraglutide alleviated CM-associated allodynia and reduced the expression of CGRP and c-fos in the TNC. We also found that liraglutide inhibited NTG-induced microglial morphological changes (process retraction) and proinflammatory factor (IL-1β and TNF-α) production in the TNC. These functions of GLP-1R in the CM model is mediated by its downstream PI3K/Akt pathway (Fig. 9). These findings provide new insights into the pathogenesis of CM and suggest that microglial GLP-1R in the TNC may be a new target for the treatment of CM. The GLP-1R agonist liraglutide, which is a commonly used hypoglycaemic drug in the clinic, might represent a new therapeutic approach for treating CM.

Acknowledgements

None.

Declarations

All animal experiments performed in this study were approved by the Ethics Committee for Animal Experimentation of Chongqing Medical University.
Not applicable.

Competing interests

The authors declare that they have no competing interests in the manuscript.
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.
Literatur
1.
Zurück zum Zitat May A, Schulte LH (2016) Chronic migraine: risk factors, mechanisms and treatment. Nat Rev Neurol 12(8):455–464PubMedCrossRef May A, Schulte LH (2016) Chronic migraine: risk factors, mechanisms and treatment. Nat Rev Neurol 12(8):455–464PubMedCrossRef
2.
Zurück zum Zitat Global regional (2018) and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392(10159):1789–1858 Global regional (2018) and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392(10159):1789–1858
3.
Zurück zum Zitat Jiang H, Deng Y, Zhang Y, Jin J, Kong X, Zhu Q, Wang K, Zhou J (2016) Field testing of the ICHD-3β and expert opinion criteria for chronic migraine. J Headache Pain 17(1):85PubMedPubMedCentralCrossRef Jiang H, Deng Y, Zhang Y, Jin J, Kong X, Zhu Q, Wang K, Zhou J (2016) Field testing of the ICHD-3β and expert opinion criteria for chronic migraine. J Headache Pain 17(1):85PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Scher AI, Stewart WF, Ricci JA, Lipton RB (2003) Factors associated with the onset and remission of chronic daily headache in a population-based study. Pain 106(1–2):81–89PubMedCrossRef Scher AI, Stewart WF, Ricci JA, Lipton RB (2003) Factors associated with the onset and remission of chronic daily headache in a population-based study. Pain 106(1–2):81–89PubMedCrossRef
6.
Zurück zum Zitat Becker WJ (2017) The Diagnosis and Management of Chronic Migraine in Primary Care. Headache 57(9):1471–1481PubMedCrossRef Becker WJ (2017) The Diagnosis and Management of Chronic Migraine in Primary Care. Headache 57(9):1471–1481PubMedCrossRef
7.
8.
Zurück zum Zitat Aurora SK, Brin MF (2017) Chronic Migraine: An Update on Physiology, Imaging, and the Mechanism of Action of Two Available Pharmacologic Therapies. Headache 57(1):109–125PubMedCrossRef Aurora SK, Brin MF (2017) Chronic Migraine: An Update on Physiology, Imaging, and the Mechanism of Action of Two Available Pharmacologic Therapies. Headache 57(1):109–125PubMedCrossRef
10.
Zurück zum Zitat Tsuda M, Inoue K, Salter MW (2005) Neuropathic pain and spinal microglia: a big problem from molecules in “small” glia. Trends Neurosci 28(2):101–107PubMedCrossRef Tsuda M, Inoue K, Salter MW (2005) Neuropathic pain and spinal microglia: a big problem from molecules in “small” glia. Trends Neurosci 28(2):101–107PubMedCrossRef
11.
Zurück zum Zitat Calvo M, Zhu N, Tsantoulas C, Ma Z, Grist J, Loeb JA, Bennett DL (2010) Neuregulin-ErbB signaling promotes microglial proliferation and chemotaxis contributing to microgliosis and pain after peripheral nerve injury. J Neurosci 30(15):5437–5450PubMedPubMedCentralCrossRef Calvo M, Zhu N, Tsantoulas C, Ma Z, Grist J, Loeb JA, Bennett DL (2010) Neuregulin-ErbB signaling promotes microglial proliferation and chemotaxis contributing to microgliosis and pain after peripheral nerve injury. J Neurosci 30(15):5437–5450PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Wendt S, Wogram E, Korvers L, Kettenmann H (2016) Experimental Cortical Spreading Depression Induces NMDA Receptor Dependent Potassium Currents in Microglia. J Neurosci 36(23):6165–6174PubMedPubMedCentralCrossRef Wendt S, Wogram E, Korvers L, Kettenmann H (2016) Experimental Cortical Spreading Depression Induces NMDA Receptor Dependent Potassium Currents in Microglia. J Neurosci 36(23):6165–6174PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Jander S, Schroeter M, Peters O, Witte OW, Stoll G (2001) Cortical spreading depression induces proinflammatory cytokine gene expression in the rat brain. J Cereb Blood Flow Metab 21(3):218–225PubMedCrossRef Jander S, Schroeter M, Peters O, Witte OW, Stoll G (2001) Cortical spreading depression induces proinflammatory cytokine gene expression in the rat brain. J Cereb Blood Flow Metab 21(3):218–225PubMedCrossRef
15.
Zurück zum Zitat Liu C, Zhang Y, Liu Q, Jiang L, Li M, Wang S, Long T, He W, Kong X, Qin G, Zhang Y, Zhou J (2018) P2 × 4-receptor participates in EAAT3 regulation via BDNF-TrkB signaling in a model of trigeminal allodynia. Mol Pain 14:1744806918795930PubMedPubMedCentral Liu C, Zhang Y, Liu Q, Jiang L, Li M, Wang S, Long T, He W, Kong X, Qin G, Zhang Y, Zhou J (2018) P2 × 4-receptor participates in EAAT3 regulation via BDNF-TrkB signaling in a model of trigeminal allodynia. Mol Pain 14:1744806918795930PubMedPubMedCentral
16.
Zurück zum Zitat Gong Q, Lin Y, Lu Z, Xiao Z (2020) Microglia-Astrocyte Cross Talk through IL-18/IL-18R Signaling Modulates Migraine-like Behavior in Experimental Models of Migraine. Neuroscience 451:207–215PubMedCrossRef Gong Q, Lin Y, Lu Z, Xiao Z (2020) Microglia-Astrocyte Cross Talk through IL-18/IL-18R Signaling Modulates Migraine-like Behavior in Experimental Models of Migraine. Neuroscience 451:207–215PubMedCrossRef
17.
Zurück zum Zitat Long T, He W, Pan Q, Zhang S, Zhang Y, Liu C, Liu Q, Qin G, Chen L, Zhou J (2018) Microglia P2 × 4 receptor contributes to central sensitization following recurrent nitroglycerin stimulation. J Neuroinflammation 15(1):245PubMedPubMedCentralCrossRef Long T, He W, Pan Q, Zhang S, Zhang Y, Liu C, Liu Q, Qin G, Chen L, Zhou J (2018) Microglia P2 × 4 receptor contributes to central sensitization following recurrent nitroglycerin stimulation. J Neuroinflammation 15(1):245PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Jing F, Zhang Y, Long T, He W, Qin G, Zhang D, Chen L, Zhou J (2019) P2Y12 receptor mediates microglial activation via RhoA/ROCK pathway in the trigeminal nucleus caudalis in a mouse model of chronic migraine. J Neuroinflammation 16(1):217PubMedPubMedCentralCrossRef Jing F, Zhang Y, Long T, He W, Qin G, Zhang D, Chen L, Zhou J (2019) P2Y12 receptor mediates microglial activation via RhoA/ROCK pathway in the trigeminal nucleus caudalis in a mouse model of chronic migraine. J Neuroinflammation 16(1):217PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Jiang L, Zhang Y, Jing F, Long T, Qin G, Zhang D, Chen L, Zhou J (2021) P2 × 7R-mediated autophagic impairment contributes to central sensitization in a chronic migraine model with recurrent nitroglycerin stimulation in mice. J Neuroinflammation 18(1):5PubMedPubMedCentralCrossRef Jiang L, Zhang Y, Jing F, Long T, Qin G, Zhang D, Chen L, Zhou J (2021) P2 × 7R-mediated autophagic impairment contributes to central sensitization in a chronic migraine model with recurrent nitroglycerin stimulation in mice. J Neuroinflammation 18(1):5PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Gong N, Xiao Q, Zhu B, Zhang CY, Wang YC, Fan H, Ma AN, Wang YX (2014) Activation of spinal glucagon-like peptide-1 receptors specifically suppresses pain hypersensitivity. J Neurosci 34(15):5322–5334PubMedPubMedCentralCrossRef Gong N, Xiao Q, Zhu B, Zhang CY, Wang YC, Fan H, Ma AN, Wang YX (2014) Activation of spinal glucagon-like peptide-1 receptors specifically suppresses pain hypersensitivity. J Neurosci 34(15):5322–5334PubMedPubMedCentralCrossRef
21.
22.
Zurück zum Zitat Campbell JE, Drucker DJ (2013) Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 17(6):819–837PubMedCrossRef Campbell JE, Drucker DJ (2013) Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 17(6):819–837PubMedCrossRef
23.
Zurück zum Zitat Athauda D, Foltynie T (2016) The glucagon-like peptide 1 (GLP) receptor as a therapeutic target in Parkinson’s disease: mechanisms of action. Drug Discov Today 21(5):802–818PubMedCrossRef Athauda D, Foltynie T (2016) The glucagon-like peptide 1 (GLP) receptor as a therapeutic target in Parkinson’s disease: mechanisms of action. Drug Discov Today 21(5):802–818PubMedCrossRef
24.
Zurück zum Zitat Hamilton A, Hölscher C (2009) Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuroreport 20(13):1161–1166PubMedCrossRef Hamilton A, Hölscher C (2009) Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuroreport 20(13):1161–1166PubMedCrossRef
25.
Zurück zum Zitat Hamilton A, Patterson S, Porter D, Gault VA, Holscher C (2011) Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain. J Neurosci Res 89(4):481–489PubMedCrossRef Hamilton A, Patterson S, Porter D, Gault VA, Holscher C (2011) Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain. J Neurosci Res 89(4):481–489PubMedCrossRef
26.
Zurück zum Zitat Abbas T, Faivre E, Hölscher C (2009) Impairment of synaptic plasticity and memory formation in GLP-1 receptor KO mice: Interaction between type 2 diabetes and Alzheimer’s disease. Behav Brain Res 205(1):265–271PubMedCrossRef Abbas T, Faivre E, Hölscher C (2009) Impairment of synaptic plasticity and memory formation in GLP-1 receptor KO mice: Interaction between type 2 diabetes and Alzheimer’s disease. Behav Brain Res 205(1):265–271PubMedCrossRef
27.
Zurück zum Zitat Liu J, Pang ZP (2016) Glucagon-like peptide-1 drives energy metabolism on the synaptic highway. FEBS J 283(24):4413–4423PubMedCrossRef Liu J, Pang ZP (2016) Glucagon-like peptide-1 drives energy metabolism on the synaptic highway. FEBS J 283(24):4413–4423PubMedCrossRef
28.
Zurück zum Zitat Zhang DG, Amin FM, Guo S, Vestergaard MB, Hougaard A, Ashina M (2020) Plasma Glucose Levels Increase During Spontaneous Attacks of Migraine With and Without Aura. Headache 60(4):655–664PubMedCrossRef Zhang DG, Amin FM, Guo S, Vestergaard MB, Hougaard A, Ashina M (2020) Plasma Glucose Levels Increase During Spontaneous Attacks of Migraine With and Without Aura. Headache 60(4):655–664PubMedCrossRef
29.
Zurück zum Zitat Won L, Kraig RP (2020) Insulin-like growth factor-1 inhibits spreading depression-induced trigeminal calcitonin gene related peptide, oxidative stress & neuronal activation in rat. Brain Res 1732:146673PubMedPubMedCentralCrossRef Won L, Kraig RP (2020) Insulin-like growth factor-1 inhibits spreading depression-induced trigeminal calcitonin gene related peptide, oxidative stress & neuronal activation in rat. Brain Res 1732:146673PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Moustafa PE, Abdelkader NF, El Awdan SA, El-Shabrawy OA, Zaki HF (2018) Liraglutide ameliorated peripheral neuropathy in diabetic rats: Involvement of oxidative stress, inflammation and extracellular matrix remodeling. J Neurochem 146(2):173–185PubMedCrossRef Moustafa PE, Abdelkader NF, El Awdan SA, El-Shabrawy OA, Zaki HF (2018) Liraglutide ameliorated peripheral neuropathy in diabetic rats: Involvement of oxidative stress, inflammation and extracellular matrix remodeling. J Neurochem 146(2):173–185PubMedCrossRef
31.
Zurück zum Zitat Deng H, Yang F, Ma X, Wang Y, Chen Q, Yuan L (2020) Long-Term Liraglutide Administration Induces Pancreas Neogenesis in Adult T2DM Mice. Cell Transplant 29:963689720927392PubMedCrossRef Deng H, Yang F, Ma X, Wang Y, Chen Q, Yuan L (2020) Long-Term Liraglutide Administration Induces Pancreas Neogenesis in Adult T2DM Mice. Cell Transplant 29:963689720927392PubMedCrossRef
32.
Zurück zum Zitat Peters CT, Choi YH, Brubaker PL, Anderson GH (2001) A glucagon-like peptide-1 receptor agonist and an antagonist modify macronutrient selection by rats. J Nutr 131(8):2164–2170PubMedCrossRef Peters CT, Choi YH, Brubaker PL, Anderson GH (2001) A glucagon-like peptide-1 receptor agonist and an antagonist modify macronutrient selection by rats. J Nutr 131(8):2164–2170PubMedCrossRef
33.
Zurück zum Zitat Sun Z, Sun L, Tu L (2020) GABAB Receptor-Mediated PI3K/Akt Signaling Pathway Alleviates Oxidative Stress and Neuronal Cell Injury in a Rat Model of Alzheimer’s Disease. J Alzheimers Dis 76(4):1513–1526PubMedCrossRef Sun Z, Sun L, Tu L (2020) GABAB Receptor-Mediated PI3K/Akt Signaling Pathway Alleviates Oxidative Stress and Neuronal Cell Injury in a Rat Model of Alzheimer’s Disease. J Alzheimers Dis 76(4):1513–1526PubMedCrossRef
34.
Zurück zum Zitat Xia J, Li Q, Liu Y, Ren Q, Gao J, Tian Y, Li J, Zhang B, Sun H, Liu S (2020) A GLP-1 Analog Liraglutide Reduces Intimal Hyperplasia After Coronary Stent Implantation via Regulation of Glycemic Variability and NLRP3 Inflammasome/IL-10 Signaling in Diabetic Swine. Front Pharmacol 11:372PubMedPubMedCentralCrossRef Xia J, Li Q, Liu Y, Ren Q, Gao J, Tian Y, Li J, Zhang B, Sun H, Liu S (2020) A GLP-1 Analog Liraglutide Reduces Intimal Hyperplasia After Coronary Stent Implantation via Regulation of Glycemic Variability and NLRP3 Inflammasome/IL-10 Signaling in Diabetic Swine. Front Pharmacol 11:372PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Zhang Y, Yuan X, Wu Y, Pei M, Yang M, Wu X, Pang Y, Wang J (2020) Liraglutide regulates bone destruction and exhibits anti-inflammatory effects in periodontitis in vitro and in vivo. J Dent 94:103310PubMedCrossRef Zhang Y, Yuan X, Wu Y, Pei M, Yang M, Wu X, Pang Y, Wang J (2020) Liraglutide regulates bone destruction and exhibits anti-inflammatory effects in periodontitis in vitro and in vivo. J Dent 94:103310PubMedCrossRef
36.
Zurück zum Zitat Gleizes C, Kreutter G, Abbas M, Kassem M, Constantinescu AA, Boisramé-Helms J, Yver B, Toti F, Kessler L (2016) β cell membrane remodelling and procoagulant events occur in inflammation-driven insulin impairment: a GLP-1 receptor dependent and independent control. J Cell Mol Med 20(2):231–242PubMedCrossRef Gleizes C, Kreutter G, Abbas M, Kassem M, Constantinescu AA, Boisramé-Helms J, Yver B, Toti F, Kessler L (2016) β cell membrane remodelling and procoagulant events occur in inflammation-driven insulin impairment: a GLP-1 receptor dependent and independent control. J Cell Mol Med 20(2):231–242PubMedCrossRef
37.
Zurück zum Zitat Pradhan AA, Smith ML, McGuire B, Tarash I, Evans CJ, Charles A (2014) Characterization of a novel model of chronic migraine. Pain 155(2):269–274PubMedCrossRef Pradhan AA, Smith ML, McGuire B, Tarash I, Evans CJ, Charles A (2014) Characterization of a novel model of chronic migraine. Pain 155(2):269–274PubMedCrossRef
38.
Zurück zum Zitat Ben Aissa M, Tipton AF, Bertels Z, Gandhi R, Moye LS, Novack M, Bennett BM, Wang Y, Litosh V, Lee SH, Gaisina IN, Thatcher GR, Pradhan AA (2018) Soluble guanylyl cyclase is a critical regulator of migraine-associated pain. Cephalalgia 38(8):1471–1484PubMedCrossRef Ben Aissa M, Tipton AF, Bertels Z, Gandhi R, Moye LS, Novack M, Bennett BM, Wang Y, Litosh V, Lee SH, Gaisina IN, Thatcher GR, Pradhan AA (2018) Soluble guanylyl cyclase is a critical regulator of migraine-associated pain. Cephalalgia 38(8):1471–1484PubMedCrossRef
39.
Zurück zum Zitat Tamborlane WV, Barrientos-Pérez M, Fainberg U, Frimer-Larsen H, Hafez M, Hale PM, Jalaludin MY, Kovarenko M, Libman I, Lynch JL, Rao P, Shehadeh N, Turan S, Weghuber D, Barrett T (2019) Liraglutide in Children and Adolescents with Type 2 Diabetes. N Engl J Med 381(7):637–646PubMedCrossRef Tamborlane WV, Barrientos-Pérez M, Fainberg U, Frimer-Larsen H, Hafez M, Hale PM, Jalaludin MY, Kovarenko M, Libman I, Lynch JL, Rao P, Shehadeh N, Turan S, Weghuber D, Barrett T (2019) Liraglutide in Children and Adolescents with Type 2 Diabetes. N Engl J Med 381(7):637–646PubMedCrossRef
40.
Zurück zum Zitat Tornehave D, Kristensen P, Rømer J, Knudsen LB, Heller RS (2008) Expression of the GLP-1 receptor in mouse, rat, and human pancreas. J Histochem Cytochem 56(9):841–851PubMedPubMedCentralCrossRef Tornehave D, Kristensen P, Rømer J, Knudsen LB, Heller RS (2008) Expression of the GLP-1 receptor in mouse, rat, and human pancreas. J Histochem Cytochem 56(9):841–851PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Farkas E, Szilvásy-Szabó A, Ruska Y, Sinkó R, Rasch MG, Egebjerg T, Pyke C, Gereben B, Knudsen LB, Fekete C (2021) Distribution and ultrastructural localization of the glucagon-like peptide-1 receptor (GLP-1R) in the rat brain. Brain struct Funct 226(1):225–245PubMedCrossRef Farkas E, Szilvásy-Szabó A, Ruska Y, Sinkó R, Rasch MG, Egebjerg T, Pyke C, Gereben B, Knudsen LB, Fekete C (2021) Distribution and ultrastructural localization of the glucagon-like peptide-1 receptor (GLP-1R) in the rat brain. Brain struct Funct 226(1):225–245PubMedCrossRef
42.
Zurück zum Zitat Xie Z, Enkhjargal B, Wu L, Zhou K, Sun C, Hu X, Gospodarev V, Tang J, You C, Zhang JH (2018) Exendin-4 attenuates neuronal death via GLP-1R/PI3K/Akt pathway in early brain injury after subarachnoid hemorrhage in rats. Neuropharmacology 128:142–151PubMedCrossRef Xie Z, Enkhjargal B, Wu L, Zhou K, Sun C, Hu X, Gospodarev V, Tang J, You C, Zhang JH (2018) Exendin-4 attenuates neuronal death via GLP-1R/PI3K/Akt pathway in early brain injury after subarachnoid hemorrhage in rats. Neuropharmacology 128:142–151PubMedCrossRef
43.
Zurück zum Zitat Alvarez E, Martínez MD, Roncero I, Chowen JA, García-Cuartero B, Gispert JD, Sanz C, Vázquez P, Maldonado A, de Cáceres J, Desco M, Pozo MA, Blázquez E (2005) The expression of GLP-1 receptor mRNA and protein allows the effect of GLP-1 on glucose metabolism in the human hypothalamus and brainstem. J Neurochem 92(4):798–806PubMedCrossRef Alvarez E, Martínez MD, Roncero I, Chowen JA, García-Cuartero B, Gispert JD, Sanz C, Vázquez P, Maldonado A, de Cáceres J, Desco M, Pozo MA, Blázquez E (2005) The expression of GLP-1 receptor mRNA and protein allows the effect of GLP-1 on glucose metabolism in the human hypothalamus and brainstem. J Neurochem 92(4):798–806PubMedCrossRef
44.
Zurück zum Zitat Scharfman HE, MacLusky NJ (2008) Estrogen-growth factor interactions and their contributions to neurological disorders. Headache 48(Suppl 2):S77–S89PubMedPubMedCentralCrossRef Scharfman HE, MacLusky NJ (2008) Estrogen-growth factor interactions and their contributions to neurological disorders. Headache 48(Suppl 2):S77–S89PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Brandes JL (2006) The influence of estrogen on migraine: a systematic review. JAMA 295(15):1824–1830PubMedCrossRef Brandes JL (2006) The influence of estrogen on migraine: a systematic review. JAMA 295(15):1824–1830PubMedCrossRef
46.
Zurück zum Zitat Chen S, Zhou M, Sun J, Guo A, Fernando RL, Chen Y, Peng P, Zhao G, Deng Y (2019) DPP-4 inhibitor improves learning and memory deficits and AD-like neurodegeneration by modulating the GLP-1 signaling. Neuropharmacology 157:107668PubMedCrossRef Chen S, Zhou M, Sun J, Guo A, Fernando RL, Chen Y, Peng P, Zhao G, Deng Y (2019) DPP-4 inhibitor improves learning and memory deficits and AD-like neurodegeneration by modulating the GLP-1 signaling. Neuropharmacology 157:107668PubMedCrossRef
47.
Zurück zum Zitat Lee CH, Jeon SJ, Cho KS, Moon E, Sapkota A, Jun HS, Ryu JH, Choi JW (2018) Activation of Glucagon-Like Peptide-1 Receptor Promotes Neuroprotection in Experimental Autoimmune Encephalomyelitis by Reducing Neuroinflammatory Responses. Mol Neurobiol 55(4):3007–3020PubMedCrossRef Lee CH, Jeon SJ, Cho KS, Moon E, Sapkota A, Jun HS, Ryu JH, Choi JW (2018) Activation of Glucagon-Like Peptide-1 Receptor Promotes Neuroprotection in Experimental Autoimmune Encephalomyelitis by Reducing Neuroinflammatory Responses. Mol Neurobiol 55(4):3007–3020PubMedCrossRef
48.
Zurück zum Zitat Liu Y, Wang L, Pan D, Li M, Li Y, Wang Y, Xu Y, Wang X, Yan J, Wu Q, Lu L, Yuan K, Yang M (2021) PET evaluation of light-induced modulation of microglial activation and GLP-1R expression in depressive rats. Transl Psychiatry 11(1):26PubMedPubMedCentralCrossRef Liu Y, Wang L, Pan D, Li M, Li Y, Wang Y, Xu Y, Wang X, Yan J, Wu Q, Lu L, Yuan K, Yang M (2021) PET evaluation of light-induced modulation of microglial activation and GLP-1R expression in depressive rats. Transl Psychiatry 11(1):26PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Hunter K, Hölscher C (2012) Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci 13:33PubMedPubMedCentralCrossRef Hunter K, Hölscher C (2012) Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci 13:33PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat McClean PL, Parthsarathy V, Faivre E, Hölscher C (2011) The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J Neurosci 31(17):6587–6594PubMedPubMedCentralCrossRef McClean PL, Parthsarathy V, Faivre E, Hölscher C (2011) The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J Neurosci 31(17):6587–6594PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Hansen HH, Barkholt P, Fabricius K, Jelsing J, Terwel D, Pyke C, Knudsen LB, Vrang N (2016) The GLP-1 receptor agonist liraglutide reduces pathology-specific tau phosphorylation and improves motor function in a transgenic hTauP301L mouse model of tauopathy. Brain Res 1634:158–170PubMedCrossRef Hansen HH, Barkholt P, Fabricius K, Jelsing J, Terwel D, Pyke C, Knudsen LB, Vrang N (2016) The GLP-1 receptor agonist liraglutide reduces pathology-specific tau phosphorylation and improves motor function in a transgenic hTauP301L mouse model of tauopathy. Brain Res 1634:158–170PubMedCrossRef
52.
Zurück zum Zitat Badawi GA, Abd El Fattah MA, Zaki HF, El Sayed MI (2017) Sitagliptin and liraglutide reversed nigrostriatal degeneration of rodent brain in rotenone-induced Parkinson’s disease. Inflammopharmacology 25(3):369–382PubMedCrossRef Badawi GA, Abd El Fattah MA, Zaki HF, El Sayed MI (2017) Sitagliptin and liraglutide reversed nigrostriatal degeneration of rodent brain in rotenone-induced Parkinson’s disease. Inflammopharmacology 25(3):369–382PubMedCrossRef
53.
Zurück zum Zitat Hakon J, Ruscher K, Romner B, Tomasevic G (2015) Preservation of the blood brain barrier and cortical neuronal tissue by liraglutide, a long acting glucagon-like-1 analogue, after experimental traumatic brain injury. PLoS One 10(3):e0120074PubMedPubMedCentralCrossRef Hakon J, Ruscher K, Romner B, Tomasevic G (2015) Preservation of the blood brain barrier and cortical neuronal tissue by liraglutide, a long acting glucagon-like-1 analogue, after experimental traumatic brain injury. PLoS One 10(3):e0120074PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Li PC, Liu LF, Jou MJ, Wang HK (2016) The GLP-1 receptor agonists exendin-4 and liraglutide alleviate oxidative stress and cognitive and micturition deficits induced by middle cerebral artery occlusion in diabetic mice. BMC Neurosci 17(1):37PubMedPubMedCentralCrossRef Li PC, Liu LF, Jou MJ, Wang HK (2016) The GLP-1 receptor agonists exendin-4 and liraglutide alleviate oxidative stress and cognitive and micturition deficits induced by middle cerebral artery occlusion in diabetic mice. BMC Neurosci 17(1):37PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Barreto-Vianna ARC, Aguila MB, Mandarim-de-Lacerda CA (2017) Beneficial effects of liraglutide (GLP1 analog) in the hippocampal inflammation. Metabo Brain Dis 32(5):1735–1745CrossRef Barreto-Vianna ARC, Aguila MB, Mandarim-de-Lacerda CA (2017) Beneficial effects of liraglutide (GLP1 analog) in the hippocampal inflammation. Metabo Brain Dis 32(5):1735–1745CrossRef
56.
Zurück zum Zitat Burstein R, Yarnitsky D, Goor-Aryeh I, Ransil BJ, Bajwa ZH (2000) An association between migraine and cutaneous allodynia. Ann Neurol 47(5):614–624PubMedCrossRef Burstein R, Yarnitsky D, Goor-Aryeh I, Ransil BJ, Bajwa ZH (2000) An association between migraine and cutaneous allodynia. Ann Neurol 47(5):614–624PubMedCrossRef
57.
Zurück zum Zitat Iyengar S, Ossipov MH, Johnson KW (2017) The role of calcitonin gene-related peptide in peripheral and central pain mechanisms including migraine. Pain 158(4):543–559PubMedPubMedCentralCrossRef Iyengar S, Ossipov MH, Johnson KW (2017) The role of calcitonin gene-related peptide in peripheral and central pain mechanisms including migraine. Pain 158(4):543–559PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Hermanson O, Telkov M, Geijer T, Hallbeck M, Blomqvist A (1998) Preprodynorphin mRNA-expressing neurones in the rat parabrachial nucleus: subnuclear localization, hypothalamic projections and colocalization with noxious-evoked fos-like immunoreactivity. Eur J Neurosci 10(1):358–367PubMedCrossRef Hermanson O, Telkov M, Geijer T, Hallbeck M, Blomqvist A (1998) Preprodynorphin mRNA-expressing neurones in the rat parabrachial nucleus: subnuclear localization, hypothalamic projections and colocalization with noxious-evoked fos-like immunoreactivity. Eur J Neurosci 10(1):358–367PubMedCrossRef
59.
Zurück zum Zitat Jones MG, Lever I, Bingham S, Read S, McMahon SB, Parsons A (2001) Nitric oxide potentiates response of trigeminal neurones to dural or facial stimulation in the rat. Cephalalgia 21(6):643–655PubMedCrossRef Jones MG, Lever I, Bingham S, Read S, McMahon SB, Parsons A (2001) Nitric oxide potentiates response of trigeminal neurones to dural or facial stimulation in the rat. Cephalalgia 21(6):643–655PubMedCrossRef
60.
Zurück zum Zitat Chen G, Zhang YQ, Qadri YJ, Serhan CN, Ji RR (2018) Microglia in Pain: Detrimental and Protective Roles in Pathogenesis and Resolution of Pain. Neuron 100(6):1292–1311PubMedPubMedCentralCrossRef Chen G, Zhang YQ, Qadri YJ, Serhan CN, Ji RR (2018) Microglia in Pain: Detrimental and Protective Roles in Pathogenesis and Resolution of Pain. Neuron 100(6):1292–1311PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Echeverry S, Shi XQ, Yang M, Huang H, Wu Y, Lorenzo LE, Perez-Sanchez J, Bonin RP, De Koninck Y, Zhang J (2017) Spinal microglia are required for long-term maintenance of neuropathic pain. Pain 158(9):1792–1801PubMedCrossRef Echeverry S, Shi XQ, Yang M, Huang H, Wu Y, Lorenzo LE, Perez-Sanchez J, Bonin RP, De Koninck Y, Zhang J (2017) Spinal microglia are required for long-term maintenance of neuropathic pain. Pain 158(9):1792–1801PubMedCrossRef
62.
Zurück zum Zitat Gu N, Eyo UB, Murugan M, Peng J, Matta S, Dong H, Wu LJ (2016) Microglial P2Y12 receptors regulate microglial activation and surveillance during neuropathic pain. Brain Behav Immun 55:82–92PubMedCrossRef Gu N, Eyo UB, Murugan M, Peng J, Matta S, Dong H, Wu LJ (2016) Microglial P2Y12 receptors regulate microglial activation and surveillance during neuropathic pain. Brain Behav Immun 55:82–92PubMedCrossRef
63.
Zurück zum Zitat Shan Y, Tan S, Lin Y, Liao S, Zhang B, Chen X, Wang J, Deng Z, Zeng Q, Zhang L, Wang Y, Hu XQ, Qiu W, Peng L, Lu ZQ (2019) The glucagon-like peptide-1 receptor agonist reduces inflammation and blood-brain barrier breakdown in an astrocyte-dependent manner in experimental stroke. J Neuroinflammation 16(1):242PubMedPubMedCentralCrossRef Shan Y, Tan S, Lin Y, Liao S, Zhang B, Chen X, Wang J, Deng Z, Zeng Q, Zhang L, Wang Y, Hu XQ, Qiu W, Peng L, Lu ZQ (2019) The glucagon-like peptide-1 receptor agonist reduces inflammation and blood-brain barrier breakdown in an astrocyte-dependent manner in experimental stroke. J Neuroinflammation 16(1):242PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Liu YY, Jiao ZY, Li W, Tian Q (2017) PI3K/AKT signaling pathway activation in a rat model of migraine. Mol Med Rep 16(4):4849–4854PubMedCrossRef Liu YY, Jiao ZY, Li W, Tian Q (2017) PI3K/AKT signaling pathway activation in a rat model of migraine. Mol Med Rep 16(4):4849–4854PubMedCrossRef
Metadaten
Titel
Activation of microglial GLP-1R in the trigeminal nucleus caudalis suppresses central sensitization of chronic migraine after recurrent nitroglycerin stimulation
verfasst von
Feng Jing
Qian Zou
Yangyang Wang
Zhiyou Cai
Yong Tang
Publikationsdatum
01.12.2021
Verlag
Springer Milan
Erschienen in
The Journal of Headache and Pain / Ausgabe 1/2021
Print ISSN: 1129-2369
Elektronische ISSN: 1129-2377
DOI
https://doi.org/10.1186/s10194-021-01302-x

Weitere Artikel der Ausgabe 1/2021

The Journal of Headache and Pain 1/2021 Zur Ausgabe

Ähnliche Überlebensraten nach Reanimation während des Transports bzw. vor Ort

29.05.2024 Reanimation im Kindesalter Nachrichten

Laut einer Studie aus den USA und Kanada scheint es bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Häusliche Gewalt in der orthopädischen Notaufnahme oft nicht erkannt

28.05.2024 Häusliche Gewalt Nachrichten

In der Notaufnahme wird die Chance, Opfer von häuslicher Gewalt zu identifizieren, von Orthopäden und Orthopädinnen offenbar zu wenig genutzt. Darauf deuten die Ergebnisse einer Fragebogenstudie an der Sahlgrenska-Universität in Schweden hin.

Fehlerkultur in der Medizin – Offenheit zählt!

28.05.2024 Fehlerkultur Podcast

Darüber reden und aus Fehlern lernen, sollte das Motto in der Medizin lauten. Und zwar nicht nur im Sinne der Patientensicherheit. Eine negative Fehlerkultur kann auch die Behandelnden ernsthaft krank machen, warnt Prof. Dr. Reinhard Strametz. Ein Plädoyer und ein Leitfaden für den offenen Umgang mit kritischen Ereignissen in Medizin und Pflege.

Mehr Frauen im OP – weniger postoperative Komplikationen

21.05.2024 Allgemeine Chirurgie Nachrichten

Ein Frauenanteil von mindestens einem Drittel im ärztlichen Op.-Team war in einer großen retrospektiven Studie aus Kanada mit einer signifikanten Reduktion der postoperativen Morbidität assoziiert.

Update AINS

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